ArticlePDF AvailableLiterature Review

Obestatin Receptor in Energy Homeostasis and Obesity Pathogenesis

Authors:

Abstract

Based on the bioinformatic prediction, Zhang and colleagues discovered obestatin, a new 23-amino acid hormone from rat stomach extract encoded by the ghrelin gene. Obestatin is present not only in the gastrointestinal tract, but also in the spleen, mammary gland, breast milk, and plasma. Obestatin appears to function as part of a complex gut-brain network whereby hormones and substances from the stomach, intestine and the brain about satiety or hunger. Given the current research regarding the effects of obestatin and its possible cognate receptor(s), this chapter provides the latest review of the physiological and pathological characteristics of this hormone and its possible receptor(s) in energy homeostasis and obesity.
Provided for non-commercial research and educational use only.
Not for reproduction, distribution or commercial use.
This chapter was originally published in the book Progress in Molecular Biology and
Translational Science, Vol. 114, published by Elsevier, and the attached copy is
provided by Elsevier for the author's benefit and for the benefit of the author's
institution, for non-commercial research and educational use including without
limitation use in instruction at your institution, sending it to specific colleagues who
know you, and providing a copy to your institution’s administrator.
All other uses, reproduction and distribution, including without limitation commercial
reprints, selling or licensing copies or access, or posting on open internet sites, your
personal or institution’s website or repository, are prohibited. For exceptions,
permission may be sought for such use through Elsevier's permissions site at:
http://www.elsevier.com/locate/permissionusematerial
From: Jian V. Zhang, Lei Li, Qingsheng Huang and Pei-Gen Ren, Obestatin Receptor
in Energy Homeostasis and Obesity Pathogenesis. In Ya-Xiong Tao, editor:
Progress in Molecular Biology and Translational Science, Vol. 114,
Burlington: Academic Press, 2013, pp. 89-107.
ISBN: 978-0-12-386933-3
© Copyright 2013 Elsevier Inc.
Academic Press
CHAPTER THREE
Obestatin Receptor in Energy
Homeostasis and Obesity
Pathogenesis
Jian V. Zhang*
,#
, Lei Li*
,1
, Qingsheng Huang*
,1
, Pei-Gen Ren
#
*Research Center for Gene and Cell Engineering, Institute of Biomedicine and Biotechnology, Shenzhen
Institute of Advance Technology, Chinese Academy of Sciences, Shenzhen, China
#
Innovative Pharmacology and Biotherapy Pre-clinical Test Public Service Platform of Shenzhen, China
1
These authors contributed equally to this work
Contents
1. Two Proposed Obestatin Receptors 90
1.1 Phylogenetic relationship of GPR39 and GLP-1R within their GPCR families 92
1.2 Gene, expression and structure of GPR39 and GLP-1R 93
1.3 Signal transduction across cell membrane for GPR39 and GLP-1R 96
2. Obestatin and its Receptor(s) in Energy Homeostasis 97
2.1 Food intake 98
2.2 Gastrointestinal motility 99
2.3 Growth hormone secretion 99
2.4 Lipid metabolism 99
2.5 GPR39 null mice 100
2.6 Obesity pathogenesis 100
Acknowledgments 102
References 102
Abstract
Based on the bioinformatic prediction, Zhang and colleagues discovered obestatin, a
new 23-amino acid hormone from rat stomach extract encoded by the ghrelin gene.
Obestatin is present not only in the gastrointestinal tract, but also in the spleen, mam-
mary gland, breast milk, and plasma. Obestatin appears to function as part of a complex
gut-brain network whereby hormones and substances from the stomach, intestine and
the brain about satiety or hunger. Given the current research regarding the effects of
obestatin and its possible cognate receptor(s), this chapter provides the latest review
of the physiological and pathological characteristics of this hormone and its possible
receptor(s) in energy homeostasis and obesity.
Progress in Molecular Biology and Translational Science, Volume 114 #2013 Elsevier Inc.
ISSN 1877-1173 All rights reserved.
http://dx.doi.org/10.1016/B978-0-12-386933-3.00003-0
89
Author's personal copy
Ghrelin stimulates food intake and adiposity and thereby increases body
weight in rodents after central as well as peripheral administration.
1
Based
on the bioinformatic prediction, Zhang and colleagues discovered that, ex-
cept ghrelin, the ghrelin gene encoded another secreted and bioactive pep-
tide. The precursor is encoded by the gene GHRL, which is located on
chromosome 3, band p25–26,
2
and is cleaved by protease into peptide seg-
ments. Zhang and colleagues isolated this new 23-amino acid hormone from
rat stomach, and it showed actions opposite to ghrelin on food intake, body
weight, and gastric emptying and named it “obestatin”.
3
In this chapter, we
summarize the possible physiological roles of obestatin and its receptor in
energy homeostasis and its possible pathological roles in obesity.
Accumulating publications have confirmed the role of obestatin in reg-
ulating energy homeostasis and its association with obesity, among many
other physiological functions. Its receptor(s) involved in these functions
remains, however, controversial. Two receptors for obestatin have been
proposed: GPR39 and GLP-1R.
3,4
Both G protein-coupled receptors
(GPCRs) are found to be correlated with the function of obestatin, especially
metabolism. Since less evidence demonstrated the exact mechanism, which
one is the innate receptor is still unknown.
5
1. TWO PROPOSED OBESTATIN RECEPTORS
GPR39 was first cloned from a fetal brain cDNA library, by hybrid-
ization at low stringency.
6
Due to the sequence similarity, GPR39, along
with another GPCR cloned at that time, GPR38, was considered related
to the growth hormone secretagogue receptor (GHS-R), whose agonists in-
cluded synthetic peptides and nonpeptide growth hormone (GH) secreta-
gogues.
6
Later the natural endogenous ligand for GHS-R was discovered,
named ghrelin,
7
and GPR39 was speculated to have a ligand of a peptide
regulating gastrointestinal functions, since all the other receptors homolo-
gous to GHS-R have such ligands.
2
GPR39 had been considered, however,
an orphan GPCR until 2005, when Zhang and colleagues discovered
obestatin encoded by the ghrelin gene. Based on the data of radiolabeled li-
gand binding assays, it was proposed that the new hormone was a cognate
ligand of GPR39.
3
The relationship of obestatin and GPR39 was later chal-
lenged by Chartrel, who stated that obestatin did not bind GPR39 and in-
duce any effects on GPR39-transfected cells.
8
In response, Zhang and
colleagues found out that the interaction of obestatin and GPR39 could
not be reproduced due to the instable bioactivity of iodinated obestatin.
9
Just
followed up their response about the bioactivity of obestatin, Zhang and
90 Jian V. Zhang et al.
Author's personal copy
colleagues found out that mono-Iodinated obestatin can bind to GPR39 and
GPR39 SNP, but not over-iodinated obestatin
13
.Duringthattime,two
groups, Holst et al. and Lauwers et al., discovered that zinc ion (Zn
2þ
) is a phys-
iologically relevant agonist of GPR39.
10,11
Both groups emphasized that
obestatin did neither bind toGPR39-expressing cellsnor display any effect such
as cAMP production. Holst et al. stated that GPR39 might signal through Gs
pathway if Zn
2þ
presented in addition to Gq and G12/13 pathways that were
constitutively activated.
10
Lauwers et al. showedthat the Zn
2þ
responsewas not
affected by obestatin. But there was still some evidence for the role of obestatin
as a GPR39 agonist. Moechars et al. constructed GPR39-knockout mice and
partially confirmed the in vivo effects of obestatin dependingon GPR39.
12
The
knockout mice’s differences in energy homeostasis included: accelerated
gastric emptying, increased gastric secretion, higher mature body weights and
body fat compositions, reduced hyperphagia after fasting, and increased choles-
terol levels. Zhang et al. demonstrated that c-fos expression was induced by
obestatin in wild-type mice but not in GPR39 null mice, suggesting that acti-
vities of obestatin depended on GPR39.
13
In addition, various studies on bone
metabolism
14
and term pregnancy
15
have shown the correlation of expression
levels of obestatin and GPR39. Thus, the role of GPR39 remains unresolved.
The other candidate receptor GLP-1R has already gained attention before
the discovery of obestatin because it is the receptor for the glucagon-like
peptide-1 (GLP-1), which highly stimulates insulin secretion and results in
antidiabetogenic effects. The cDNA of GLP-1R was first isolated from a pan-
creatic islet cDNA library from rats, and its function as a receptor for GLP-1
was confirmed by binding experiments with radiolabeled GLP-1.
16
The
cDNA of GLP-1R of human was isolated by several groups in 1993.
17–19
At the same time, two exogenous ligands were identified, which were
exendin-4 (Ex-4) and exendin-(9–39).
19
The interation of GLP-1R to
obestatin was revealed by Granata et al.in2008.
4
They observed that obestatin
bound to GLP-1R and promoted b-cell and islet cell survival. The binding
of radiolabeled GLP-1 to the cell membrane was slightly inhibited by motilin,
GLP-1, acylated ghrelin, [D-Lys
3
]-GHRP-6, and Ex-9, and obestatin
dose-dependently inhibited the binding of GLP-1 to the cell membrane.
4
Thus, the binding assay indicated that obestatin and GLP-1 recognized the
same binding sites.
4
Furthermore, obestatin did not influence GLP-1 secre-
tion, thus the promotion of cell survival was independent of GLP-1.
4
Favaro
et al. later confirmed that the anti-apoptotic effects of obestatin and exendin-4
involved PI3K/Akt, ERK1/2, and cAMP/PKA pathways.
20
Astudyon
the metabolic effects of obestatin in rats yielded several negative results
that obestatin neither bound to cells overexpressing GLP-1R, nor altered
91Obestatin Receptor in Energy Homeostasis and Obesity Pathogenesis
Author's personal copy
insulin release from islets, where both GLP-1R and GPR39 should be
expressed.
21
These controversies on both the obestatin receptor and its related
biological functions highlight the urgency of the introduction of study aspects
other than cell biology and physiology.
1.1. Phylogenetic relationship of GPR39 and GLP-1R
within their GPCR families
GPR39 and GLP-1R are both GPCRs. GPCR is a seven-transmembrane
protein, with a N-terminal extracellular region and three extracellular loops
recognizing a ligand molecule and a C-terminal intracellular region and
three intracellular loops interacting with a G protein. Although all GPCRs
possess this structural feature, they are results of convergent evolution.
GPR39 and GLP-1R belong to two different families sharing little sequence
similarity.
GPR39 is characterized as a member of family A, or rhodopsin-like
GPCR. The sequence of GPR39 is most related to the ghrelin receptor
(or GHS-R), the neurotensin receptors, the motilin receptor (GPR38),
and neuromedin-U receptors.
6
The ghrelin receptor is the cognitive recep-
tor for the peptide hormone ghrelin, which is encoded by the same gene of
obestatin but with opposite roles to obestatin in the control of food intake.
22
The two neurotensin receptors, NTS-R1 and NTS-R2, are receptors of
a tridecapeptide neurotensin.
23,24
Neurotensin is a cleavage segment of a
17 kDa precursor protein, which is expressed in the brain and gut.
24
Neu-
rotensin and its two receptors are correlated to the regulation of dopaminer-
gic neurotransmissions.
24
The motilin receptor was once known as GPR38,
cloned at the same time of GPR39.
6,25
Its ligand, the motilin peptide,
regulates interdigestive gastrointestinal motility by stimulating rhythmic
contraction of the smooth muscle.
25
The two neuromedin-U receptors
are NMU-R1 and NMU-R2. Neuromedin-U, similar to motilin, induces
smooth muscle contraction. NUM-R1 is expressed in greatest abundance
in the gastrointestinal and urogenital systems but at a much lower
level within the central nervous system, and NUM-R2 is expressed in a
complementary manner, lower in peripheral tissues but higher in the central
nervous system.
26
GLP-1R belongs to GPCR family B, or the secretin-like family, charac-
terized by the three disulfide bonds in the N-terminal domain.
27
Ligands for
receptors in this family are large peptides with paracrine activities.
28
By
sequence similarity, GLP-1R forms a subgroup with GLP-2R, GCGR
92 Jian V. Zhang et al.
Author's personal copy
(the glucagon receptor), and GIPR (the gastric inhibitory polypeptide
receptor).
28
These receptors, together with their ligands, are all essential in
maintaining glucose homeostasis. GLP-2R is stimulated by GLP-2, a
33-amino acid peptide cleaved from proglucagon,
29
which also encodes gluca-
gon and GLP-1. GLP-2R does not respond to GLP-1.
29
GCGR recognizes
glucagon and plays a role in controlling the production of hepatic glucose
and insulin secretion, so as to regulate the level of blood glucose.
30
GIPR is a
receptor for GIP,
31
a hormone that stimulates insulin secretion and thus is a
potential drug treating type 2 diabetes.
32
1.2. Gene, expression and structure of GPR39 and GLP-1R
The human GPR39 gene is located in chromosome 2, band q21–q22,
6
con-
sisting of two exons.
33
The first exon encodes residues 1 to 285, corres-
ponding to the N-terminal region and the first five transmembrane
helices, and the second exon encodes residues 286 to 435, corresponding
to the last two transmembrane helices and the C-terminal region.
34
Between
the two exons there is an intron as large as 200 kb, whose analogue appears in
the genes of the ghrelin receptor and the motilin receptor.
34
The second
exon of GPR39 shares a segment of DNA with LYPD1 gene
34
: GPR39
is encoded by the forward strand of DNA, while LYPD1 is transcribed from
the reverse strand. The two-exon structure and LYPD1 gene-overlapping
pattern are conserved among several species.
34
Beside the canonical form
of GPR39 (named GPR39-1a), which is 435-amino acid long and has
seven-transmembrane helices,
6
a splice variant GPR39-1b has been identi-
fied, which is encoded by the first exon and thus truncated after the fifth
transmembrane helix.
34
The maximal promoter activity resides in the 50-UTR region. Such a re-
gion in U-138 cell lines was reported to be from 673 to 14 from the trans-
lation initiation codon, while in HepG2 cell from 573 to 14.
34
Although
the negative regulatory elements have not yet been determined, there was
such an element from 673 to 573 in HepG2 cells. The translation initia-
tion codon is at 469 nucleotides downstream of the transcriptional start site.
34
As a product of transcription, alternative splicing, and translation, full-
length GPR39-1a is a protein with a molecular weight of 52 kDa
15
; another
product, the GPR39-1b protein, with a molecular weight of 32 kDa, is
identical in sequence to GPR39-1a for residues 1 to 285, and after residue
285, its sequence is Ser-Glu-Ser and end, which is different from the
corresponding sequence of GPR39-1a.
35
93Obestatin Receptor in Energy Homeostasis and Obesity Pathogenesis
Author's personal copy
GPR39 is expressed in many organs and various tissues, especially in
stomach, white adipose tissue, and hypothalamus.
6,34,36
GPR39-1b is
widely expressed but may be nonfunctional. Egerod et al. pointed out that
some studies before might have been misled by the unawareness of GPR39-
1b and LYPD1. Both have a region of sequence overlapping with GPR39-
1a, the canonical and functional receptor.
34
The level of constitutive activity
of GPR39-1a could be affected by the splicing producing GPR39-1b,
which may be an implication of the conservation of the two-exon structure
and LYPD1 gene-overlapping pattern.
34
Furthermore, there are GPCRs
that have splice variants like GPR39. For the luteinizing hormone receptor,
the calcitonin receptor, and a1A-adrenoceptor, the variants with shorter
sequences interact, perhaps by forming heterodimers, with the full-length
receptors and alter their functions.
35
Because GPR39, similar to related
GPCRs such as the ghrelin receptor, keeps a rather high constitutive,
ligand-independent activity, the expression of GPR39-1b may be a way
to change the signaling of GPR39-1a.
The human GPL-1R gene has been mapped to chromosome 6, band
p21.1,
37
including 13 exons.
38
Along with the gene, there are two regions
with highly polymorphic simple sequence repeat, named CA1 and CA3.
39
The simple tandem repeat DNA polymorphism adopts the form of (TG)
n
37
and discriminates many alleles. When these two repeats were identified,
14 alleles for CA1 were observed in African-Americans and six in Cauca-
sians, and 16 alleles for CA3 were observed in African-Americans and eight
in Caucasians.
39
Various analyses of allelic frequencies in noninsulin-
dependent diabetes mellitus patients negate the hypothesis that inherited
defects in GLP-1R in African-Americans and Caucasians determine the
genetic susceptibility to noninsulin-dependent diabetes mellitus.
39
In the 50-UTR of the GLP-1R gene, three binding sites for transcript
factor Sp1 have been localized.
40
By experiments with constructed mutants,
mutation of the two proximal sites, 123 and 58 nucleotides upstream from
the transcription start site, were shown to be relevant to the reduction of
activity, while the distal one, 339 nucleotides upstream from the transcrip-
tion start site, to be a cis-regulatory-element as a repressor. Both Sp1 and Sp3
could bind to these sites, involved in the regulation of the expression.
40
Be-
sides, there is yet another distal cell-specific silencer element. This element
lies between 2285 to 2235 in the 50-flanking region of the GLP-1R
gene. The silencer element may function for tissue- and cell-specific expres-
sion of GLP-1R, because Sp1 is a ubiquitous transcription factor
maintaining basal promoter activity. It was reported that the transcription
94 Jian V. Zhang et al.
Author's personal copy
of the human GLP-1R gene was repressed in fibroblasts and pancreatic
A-cells, and not pancreatic B- and D-cells.
41
Structural study on GPCR family B is less bustling than that on GPCR
family A. There are no experimentally determined structures of full-length
receptors of family B, but structures of the N-terminal extracellular domains
of several receptors, including GLP-1R and GIPR. These structures reveal
detailed insights of the receptors, for example, the structural mechanism of
the recognition between ligands and receptors. In the crystal structure of
GPL-1R extracellular domain bound with an antagonist exendin-4
(9–39),
27
the N-terminal domain of GLP-1R embraces the antagonist pep-
tide, forming an amphipathic a-helix, by both hydrophobic and hydrophilic
interactions. Similar to other solved structures of GPCR family B, the core of
the N-terminal domain of GPL-1R possesses three conserved disulfide bonds
and two regions of antiparallel b-sheets.
27
The hydrophobic binding site is
defined by several a-helices and a loop linking two antiparallel b-strands.
In contrast to a hypothesis
42
suggested by experiments with truncated ana-
logues of exendin-4 and explaining the superior affinity of exendin-4, the
Trp cage of exendin-4, which is a segment of nine amino acid residues in
the C-terminus of exendin-4, does not participate the in interaction with
GLP-1R.
27
The binding pattern of exendin-4 with GLP-1R is similar to that
of other GPCRs of family B with their ligands, for example, GIPR and GIP.
43
The crystal structure displaying the binding pattern confirms a two-step model
of the binding mechanism
44
: The first step is the C-terminal region of the pep-
tide ligand interacts with the N-terminal extracellular domain of the GPCR,
adjusting the orientation and may be secondary structure; the second step is
the N-terminal region of the ligand touches the other part of the GPCR, es-
pecially the transmembrane helices, leading to signal transduction. Structure of
the other portion of GLP-1R is unknown and believed similar to GPCR of
family A.
43
There is still the forth disulfide bond formed by residue Cys226
and Cys296, in which Cys296 belongs to the third extracellular helix and
is close to the activation pocket because mutation of Cys226 to alanine de-
creases GLP-1 potency while mutation of Cys296 to alanine in addition re-
stores the lost potency.
45
In the 463-amino acid-long sequence, residues
Cys341 and Arg348 are conserved and could be sites of mono-ADP-
ribosylation.
46
GLP-1R was first known as a regulator of insulin secretion and mainly
expressed in pancreatic b-cells. In other tissue, GLP-1R mRNA can be
identified in lung, stomach, kidney, hypothalamus, brain, and heart, but
not in skeletal muscle, liver, and adipose.
47,48
95Obestatin Receptor in Energy Homeostasis and Obesity Pathogenesis
Author's personal copy
1.3. Signal transduction across cell membrane for GPR39
and GLP-1R
GPCR mediates signaling by association with G proteins and involves sec-
ond messengers such as cAMP and calcium ions (Ca
2þ
). Some signaling pro-
cess of GPR39 might be triggered by obestatin or zinc ions, beside pathways
that are thought to be constitutively activated.
10
The obestatin-binding sites
have not yet been identified, although a Gs-mediated signaling pathway has
been proposed.
3
In cultured preadipocytes, obestatin bound to GPR39 is
reported to stimulate the phosphorylation of ERK1/2 and potentially
Akt, the downstream signaling partners.
4,13
Obestatin induces the
expression of c-fos, a transcription factor as a marker of activity in gastroin-
testinal and other tissues. If tested with GPR39-null mice, the obestatin-
induced c-fos expression was abolished, confirming the role of GPR39 in
inducing the action of obestatin.
13
The zinc ion-binding sites are residues
His17 and His19 according to mutagenesis experiments.
49
If Zn
2þ
is present,
GPR39 might also signal through Gs pathway, in addition to Gq and
G12/13 pathways that are constitutively activated.
10
It remains unknown,
however, if Zn
2þ
is only an allosteric modulator enhancing the function
of another hormone.
10
GLP-1R is a GPCR for GLP-1,
17
a hormone playing a key role in
incretin effect. The term “incretin” is used to denote the fact that a plasma
insulin response can be greater to oral glucose load than to an isoglycemic
intravenous glucose infusion.
50
Incretin effect is impeded in patients with
type 2 diabetes, thus incretin-based therapies, for example, with GLP-1R
agonists such as Liraglutide (approved by Europe in 2009), seem promising
for the treatment of the disease.
51
By analysis of mice with null mutations,
it was demonstrated that loss of GLP-1R action results in impaired insulin
response to oral glucose.
52
GLP-1R couples to Gs, Gq/11, and Gi1/2.
53
Via the coupling, it can trigger the activation of phospholipase C
54
and
MAPK,
53
as well as adenylyl cyclase,
55
leading to increases of cytosolic cal-
cium. The Lys334-Leu335-Lys336 sequence within the third intracellular
loop is a putative site for G protein coupling, for deletion of the sequence
abolishes the activation of adenylyl cyclase and insulin secretion.
56–58
In
this loop, residues V327, I328, V331, and K334 may be also essential in
interaction with G protein and stimulation of cAMP production, as shown
in an alanine scanning experiment.
59
These residues were located in one
face of an a-helix in a helical wheel model, suggesting that the secondary
structure of the third intracellular region consists of an a-helix.
59
In studies
96 Jian V. Zhang et al.
Author's personal copy
with synthetic peptides analogous to this region, the peptide of the entire
loop strongly activated pertussis toxin- and cholera toxin-sensitive G pro-
teins.
58
By measurements of GTPase activity stimulated by these ana-
logues, the effect of the third intracellular loop was further divided into
two parts: the N-terminal half for activating cholera toxin-sensitive G pro-
teins (Gs), and the C-terminal half for activating pertussis toxin-sensitive G
proteins (Gi).
58
This division provides an explanation of the existence of
several G protein partners of GLP-1R.
The interaction of obestatin and GLP-1R was first identified by the
transduction of survival signals.
4
In the study, obestatin showed specific
binding on HIT-T15 cells and INS-1E b-cells, both of which were from
islet b-cells and induced the proliferation and survival of serum-starved
and cytokine-treated cells. With the treatment of obestatin, intracellular
cAMP concentrations in INS-1E cell increased in 30 min and then decreased
but still were higher than control at 60 and 90 min.
4
Any one of the antag-
onist of the Gs protein, inhibitor of adenylyl cyclase, and inhibitor of protein
kinase A blocked the anti-apoptotic effect induced by obestatin. In addition,
obestatin promoted ERK1/2 and Akt phosphorylation but not Src kinase,
suggesting that the anti-apoptotic effect of obestatin on b-cells depended on
adenylyl cyclase/cAMP/PKA, PI3K/Akt, and ERK1/2.
4
2. OBESTATIN AND ITS RECEPTOR(S) IN ENERGY
HOMEOSTASIS
Two independent studies revealed obestatin immunoreactivity in cells of
the gastric mucosa, myenteric plexus, and in Leydig cells of thetestis
60
and in the
stomach,duodenum, jejunum, colon, and pancreas
61
of Sprague–Dawley rats,
respectively. The obestatin immunoreactivity in human tissue was localized to
the mucosa of the gastrointestinal tract, the pancreatic islets, and the epithelial
cells in the ducts of mammary glands.
62
The GPR39 immunoreactivity was
localized to gastric mucosa, intestinal villi, white adipose tissues, hepatic cords,
and kidney tubules of adult mice.
13
The colocalization of immunoreactive
obestatin and its receptor GPR39 in the gastric mucosa and intestinal tissues
implies its involvement in energy homeostasis.
Obestatin may play a role in energy homeostasis through different phys-
iological processes, like modulating food intake and gastrointestinal motility,
GH secretion, and lipid metabolism.
97Obestatin Receptor in Energy Homeostasis and Obesity Pathogenesis
Author's personal copy
2.1. Food intake
In the first report of obestatin, Zhang et al. showed that the intraperitoneal
injection (125 and 1000 nmol/kg) and intracerebroventricular treatment
(8 nmol/kg) of obestatin in adult male mice decreased food intake.
3
Re-
sults also showed that treatment with obestatin for 5, 6, 7, and 8 days
(1000 nmol/kg, three times a day) in adult male rats resulted in suppressed
body weight gain.
3
Then many groups repeated its anorectic effects in a
number of experimental paradigms after both peripheral and central ad-
ministration of obestatin. Bresciani et al. showed that obestatin inhibited
feeding after short-term starvation in young adult male rats.
63
Green
et al. reported a 43% reduction in food intake after intraperitoneal obestatin
injection 4 h prior to an allowed 15 min period of feeding in mice.
64
Bru-
netti et al. also repeated its anorectic effect by intraperitoneal daily injection
of obestatin for 12 days. The decrease in food intake and weight gain be-
came evident on day 6 and day 8, respectively, and maintained through to
day 12.
65
Sibilia et al. reported that acute intracerebroventricular admin-
istration of obestatin in 16 h-starved rats induced a trend toward a reduc-
tion of food intake but not statistically significant. Chronic
intracerebroventricular administration of obestatin for 1 day significantly
decreased food intake but this effect vanished thereafter for the next
2–28 days.
66
Based on their study, Sibilia et al. concluded that the effect
of obestatin on food intake tends to be short-term instead of long-term
influence.
66
Since administration of ghrelin induces potent stimulating effects on
food intake, the relation between obestatin and ghrelin in food intake
was also studied. Though Zizzari et al. found no difference in the food intake
at 1, 3, 5, and 18 h postintraperitoneal injection of 1000 nmol/kg obestatin
in either 24 h fasted mice or normal diet mice, they showed that obestatin
inhibited ghrelin-induced orexigenic effect in normal diet mice.
67
On the other hand, several researchers were not able to confirm the effect
of obestatin on food intake,
21,68–71
thus the role of obestatin as an anorectic
hormone was challenged.
72
The reason for the discrepancies is unclear. The
different experimental paradigms such as different animal species, methods
of administration, and the duration of drug treatment, might have influenced
the outcome of the effect of obestatin on food intake in some way. Also, a
recent study questioning the purity of synthetic peptides including obestatin
might shed a clue on these discrepancies.
73
98 Jian V. Zhang et al.
Author's personal copy
2.2. Gastrointestinal motility
Not only food intake Zhang et al. also showed that jejunal contraction was
inhibited and obestatin treatment resulted in a sustained suppression of gas-
tric emptying activity.
3
Ataka et al. showed that intravenous injection of
obestatin inhibited motor activity in the stomach antrum and duodenum
of freely moving conscious rats and that CRF type 1 and type 2 receptors
in the brain might be involved in these effects of obestatin on gastroduodenal
motility.
74
These results were reproduced in another study using freely
moving conscious rat models.
75
Some other studies also addressed the effect of obestatin on ghrelin-
induced gastrointestinal motility. Depoortere et al. reported that peripheral
administration of obestatin showed no effect on fundic strip contractions but
cotreatment of obestatin tended to reduce the excitatory effect of ghrelin in
mice.
76
However, another study reported that obestatin failed to antagonize
ghrelin-induced stimulation of gastroduodenal motility.
74
In summary, the effect of obestatin on gastrointestinal motility remains
controversial. The effects of obestatin in gastric emptying and the contrac-
tility of the strips from fundus and small intestine of mice and rats were not
reproduced by De Smet et al..
77
2.3. Growth hormone secretion
Zhang et al. showed that unlike ghrelin, obestatin did not increase in vitro GH
secretion by cultured rat pituitary cells,
3
which was confirmed by other
groups. Bresciani et al. showed that obestatin had no effect on GH secretion
or GH-stimulated hexarelin production or in spontaneous corticosterone se-
cretion in 10-day-old rats.
63
Nogueiras et al. reported no effect of obestatin
treatment not only on GH release but also some hypothalamic neuropeptides
involved in energy balance regulation.
68
However, Zizzari et al. reported
obestatin (10 mg/rat)-affected ghrelin-induced GH secretion in vivo though
obestatin did not affect spontaneous or ghrelin-induced GH release from
superfused pituitaries ex vivo or basal GH levels in vivo.
67
2.4. Lipid metabolism
Another physiological role of obestatin might be as a regulator of adipocyte
metabolism and adipogenesis. Zhang and colleagues showed that treatment
of preadipocytes 3T3-L1 cells with obestatin induced an early-response
gene c-fos and stimulated ERK1/2 phosphorylation.
13
Then the study by
99Obestatin Receptor in Energy Homeostasis and Obesity Pathogenesis
Author's personal copy
Gurriara
´n-Rodrı
´guez et al. supported a role of obestatin in promoting
adipogenesis and adipocyte metabolism regulation.
78
Obestatin activated Akt
and its downstream targets while it inactivated AMPK and decreased ACC
phosphorylation in 3T3-L1 cells. Obestatin treatment together with IBMX
and DEX was shown to regulate the expression of C/EBPa,C/EBPb,C/EBPd,
and PPARgpromoting adipogenesis. The involvement of obestatin in cell
metabolism and adipogenesis was confirmed in vivo in the white adipose tissue
obtained after 24 and 72 h continuous subcutaneous infusion of obestatin.
78
Miegueu et al. reported that obestatin stimulated cell differentiation and increased
fatty acid uptake in a dose-dependent manner while obestatin decreased lipolysis
in 3T3-L1 cells.
79
in vivo study showed that plasma triglyceride levels were
significantly reduced by a chronic 14-day treatment with a stable obestatin
analog, N-terminally PEGylated obestatin.
80
Cholesterol levels were increased
significantly in the putative obestatin receptor GPR39 null mice compared with
wild-type mice.
12
These results support a role of obestatin in lipid metabolism.
2.5. GPR39 null mice
GPR39 knockout mice may reveal the physiological roles of obestatin/
GPR39. Moechars et al. showed that GPR39 null mice showed accelerated
gastric emptying, increased volume of gastric secretion, higher mature body
weight and body fat composition, and increased cholesterol levels compared
with wild type.
12
The report also showed that food intake during day 1 was
not different between the two genotypes while food intake after fasting was
decreased in GPR39 null mice.
12
Then Zhang and colleagues further
showed that treatment with obestatin induced an early-response gene c-fos
expression in gastric mucosa of wild-type, but not GPR39 null, mice, und-
erscoring a mediating role of this receptor in obestatin actions.
13
The altered
gastric activity, body weight, body fat composition, and cholesterol levels in
GPR39 null mice might be due to the lost of obestatin biological function
due to its missing receptor. All these supported a role of obestatin-GPR39
system in energy homeostasis.
2.6. Obesity pathogenesis
Obesity with its increasing prevalence becomes a global epidemic problem.
In the United States, more than 20% of adults are clinically obese as defined
by a body mass index of 30 kg/m
2
or higher, and an additional 30% are over-
weight.
81
As a new proposed anorectic peptide hormone, the association of
obestatin with obesity and obesity-related diseases draws great interest ever
100 Jian V. Zhang et al.
Author's personal copy
since its discovery. Considering the involvement of both ghrelin and
obestatin in energy homeostasis, both the plasma obestatin level and plasma
ghrelin/obestatin ratio were used to assess the possible involvement of
obestatin in obesity pathogenesis. Several groups reported decreased
plasma obestatin level in obese adults than normal weight control
15,82–86
among which Guo et al. detected an increased ghrelin to obestatin ratio
and Huda et al. detected no difference in ghrelin to obestatin ratio between
obese and lean subjects
1
accompanied by decreased obestatin level. On
the contrary, Vicennati et al. reported a higher obestatin level in obese
women compared to control together with a decreased ghrelin to obestatin
ratio.
87
Zou et al. reported no significant difference in plasma obestatin
level between obese and normal weight children with a decreased ghrelin
to obestatin ratio.
88
Due to the heterogeneity in the relationship between plasma obestatin
levels and obesity, Zhang et al. performed a meta-analysis on the related stud-
ies with obestatin hormone level with the published work before December
2009.
89
Concerning obestatin levels in the nine studies with 566 participants,
obestatin of the obese groups is lower than that of the normal weight groups
at 64.19 pg/mL [95% confidence interval (CI) ¼36.11–92.26, P<0.01].
Concerning the ghrelin/obestatin ratio in the five studies with 259 partici-
pants, the ratios of obese group were lower than those of the normal weight
group although not statistically significant.
89
Not only the plasma obestatin level was showed to be associated with
obesity, but gastric body mucosa obestatin expression was also reported to
decrease in overweight and obese patients by immunohistochemistry.
90
The association between obestatin levels (both circulating and gastric level)
with obesity all suggested a possible role for stable obestatin level or balanced
obestatin/ghrelin ratio in long-term body weight regulation.
The effects of weight loss surgery or exercise on obestatin level might
give us another perspective to decipher the relationship between obestatin
and obesity. Changes in plasma obestatin levels after weight loss through
either Roux-en-Y gastric bypass (RYGB) surgery or weight reduction in
summer camp were studied. Roth et al. reported that ghrelin concentrations
and ghrelin/obestatin ratios were lower in patients 2 years after RYGB
surgery than before, while obestatin levels did not change.
91
Martins et al.
reported that both ghrelin and obestatin plasma levels were significantly
elevated in the patients 3 years after the RYGB surgery as compared to the
control group and no statistically significant difference in ghrelin/obestatin ra-
tio was detected among the two groups.
92
In the study by Huda et al.,
101Obestatin Receptor in Energy Homeostasis and Obesity Pathogenesis
Author's personal copy
obestatin tended to be lower in gastrectomy subjects and their obestatin/
ghrelin ratio differed from healthy controls.
86
A 24 h to 7-day time-course
study on the effect of short-term anaerobic exercise training was performed
in college women though no effect of the exercise on the circulating obestatin
levels was observed.
93
Accumulating evidence piled up for the involvement of obestatin in
energy homeostasis and its association with obesity. But more investigation
is needed to decipher the detailed mechanism of its physiological and path-
ological roles including the innate receptor(s) for obestatin, from which
we can understand more about the regulation of energy homeostasis and
hopefully provide candidate(s) for obesity-related treatment.
ACKNOWLEDGMENTS
This work was supported by the following grants: One-hundred Talent Program of
Chinese Academy of Sciences (to J. V. Z.), The National Basic Research Program of
China (973 program 2013CB945503), Distinguished Young Scholars Program of The
National Natural Science Foundation of China (NSFC 31100850 to J. V. Z.), The
Distinguished Young Scholars of Shenzhen City (SW201110059 to J. V. Z.), Shenzhen
Engineering Laboratory of Single-molecule Detection and Instrument Development
and Innovative Pharmacology and Biotherapy Pre-clinical Test Public Service Platform,
Shenzhen.
REFERENCES
1. Inui A. Ghrelin: an orexigenic and somatotrophic signal from the stomach. Nat Rev
2001;2:551–60.
2. Kojima M, Kangawa K. Ghrelin: structure and function. Physiol Rev 2005;85:495–522.
3. Zhang JV, Ren PG, Avsian-Kretchmer O, Luo CW, Rauch R, Klein C, et al. Obestatin,
a peptide encoded by the ghrelin gene, opposes ghrelin’s effects on food intake. Science
2005;310:996–9.
4. Granata R, Settanni F, Gallo D, Trovato L, Biancone L, Cantaluppi V, et al. Obestatin
promotes survival of pancreatic beta-cells and human islets and induces expression of
genes involved in the regulation of beta-cell mass and function. Diabetes 2008;
57:967–79.
5. Lacquaniti A, Donato V, Chirico V, Buemi A, Buemi M. Obestatin: an interesting but
controversial gut hormone. Ann Nutr Metab 2011;59:193–9.
6. McKee KK, Tan CP, Palyha OC, Liu J, Feighner SD, Hreniuk DL, et al. Cloning and
characterization of two human G protein-coupled receptor genes (GPR38 and GPR39)
related to the growth hormone secretagogue and neurotensin receptors. Genomics
1997;46:426–34.
7. Kojima M, Hosoda H, Matsuo H, Kangawa K. Ghrelin: discovery of the natural endog-
enous ligand for the growth hormone secretagogue receptor. Trends Endocrinol Metab
2001;12:118–22.
8. Chartrel N, Alvear-Perez R, Leprince J, Iturrioz X, Reaux-Le Goazigo A, Audinot V,
et al. Comment on “Obestatin, a peptide encoded by the ghrelin gene, opposes ghrelin’s
effects on food intake” Science 2007;315:766 [author reply 766].
102 Jian V. Zhang et al.
Author's personal copy
9. Zhang JV, Klein C, Ren PG, Kass S, Donck LV, Moechars D, et al. Response to Com-
ment on “Obestatin, a peptide encoded by the ghrelin gene, opposes ghrelin’s effects on
food intake” Science 2007;315:766.
10. Holst B, Egerod KL, Schild E, Vickers SP, Cheetham S, Gerlach LO, et al. GPR39
signaling is stimulated by zinc ions but not by obestatin. Endocrinology 2007;148:13–20.
11. Lauwers E, Landuyt B, Arckens L, Schoofs L, Luyten W. Obestatin does not activate
orphan G protein-coupled receptor GPR39. Biochem Biophys Res Commun 2006;
351:21–5.
12. Moechars D, Depoortere I, Moreaux B, de Smet B, Goris I, Hoskens L, et al. Altered
gastrointestinal and metabolic function in the GPR39-obestatin receptor-knockout
mouse. Gastroenterology 2006;131:1131–41.
13. Zhang JV, Jahr H, Luo CW, Klein C, Van Kolen K, Ver Donck L, et al. Obestatin
induction of early-response gene expression in gastrointestinal and adipose tissues and
the mediatory role of G protein-coupled receptor, GPR39. Mol Endocrinol 2008;22:
1464–75.
14. Pacheco-Pantoja EL, Ranganath LR, Gallagher JA, Wilson PJ, Fraser WD. Receptors
and effects of gut hormones in three osteoblastic cell lines. BMC Physiol 2011;11:12.
15. Fontenot E, DeVente JE, Seidel ER. Obestatin and ghrelin in obese and in pregnant
women. Peptides 2007;28:1937–44.
16. Thorens B. Expression cloning of the pancreatic beta cell receptor for the gluco-incretin
hormone glucagon-like peptide 1. Proc Natl Acad Sci USA 1992;89:8641–5.
17. Dillon JS, Tanizawa Y, Wheeler MB, Leng XH, Ligon BB, Rabin DU, et al. Cloning
and functional expression of the human glucagon-like peptide-1 (GLP-1) receptor.
Endocrinology 1993;133:1907–10.
18. Graziano MP, Hey PJ, Borkowski D, Chicchi GG, Strader CD. Cloning and functional
expression of a human glucagon-like peptide-1 receptor. Biochem Biophys Res Commun
1993;196:141–6.
19. Thorens B, Porret A, Buhler L, Deng SP, Morel P, Widmann C. Cloning and functional
expression of the human islet GLP-1 receptor. Demonstration that exendin-4 is an ag-
onist and exendin-(9–39) an antagonist of the receptor. Diabetes 1993;42:1678–82.
20. Favaro E, Granata R, Miceli I, Baragli A, Settanni F, Cavallo Perin P, et al. The ghrelin
gene products and exendin-4 promote survival of human pancreatic islet endothelial cells
in hyperglycaemic conditions, through phosphoinositide 3-kinase/Akt, extracellular
signal-related kinase (ERK)1/2 and cAMP/protein kinase A (PKA) signalling pathways.
Diabetologia 2012;55:1058–70.
21. Unniappan S, Speck M, Kieffer TJ. Metabolic effects of chronic obestatin infusion in
rats. Peptides 2008;29:1354–61.
22. Kojima M, Hosoda H, Date Y, Nakazato M, Matsuo H, Kangawa K. Ghrelin is a
growth-hormone-releasing acylated peptide from stomach. Nature 1999;402:656–60.
23. Vita N, Laurent P, Lefort S, Chalon P, Dumont X, Kaghad M, et al. Cloning and
expression of a complementary DNA encoding a high affinity human neurotensin recep-
tor. FEBS Lett 1993;317:139–42.
24. Vita N, Oury-Donat F, Chalon P, Guillemot M, Kaghad M, Bachy A, et al. Neurotensin
is an antagonist of the human neurotensin NT2 receptor expressed in Chinese hamster
ovary cells. Eur J Pharmacol 1998;360:265–72.
25. Feighner SD, Tan CP, McKee KK, Palyha OC, Hreniuk DL, Pong SS, et al. Receptor
for motilin identified in the human gastrointestinal system. Science 1999;284:2184–8.
26. Raddatz R, Wilson AE, Artymyshyn R, Bonini JA, Borowsky B, Boteju LW, et al.
Identification and characterization of two neuromedin U receptors differentially
expressed in peripheral tissues and the central nervous system. J Biol Chem 2000;275:
32452–9.
103Obestatin Receptor in Energy Homeostasis and Obesity Pathogenesis
Author's personal copy
27. Runge S, Thogersen H, Madsen K, Lau J, Rudolph R. Crystal structure of the ligand-
bound glucagon-like peptide-1 receptor extracellular domain. J Biol Chem 2008;283:
11340–7.
28. Fredriksson R, Lagerstrom MC, Lundin LG, Schioth HB. The G-protein-coupled
receptors in the human genome form five main families. Phylogenetic analysis, para-
logon groups, and fingerprints. Mol Pharmacol 2003;63:1256–72.
29. Munroe DG, Gupta AK, Kooshesh F, Vyas TB, Rizkalla G, Wang H, et al. Prototypic
G protein-coupled receptor for the intestinotrophic factor glucagon-like peptide 2. Proc
Natl Acad Sci USA 1999;96:1569–73.
30. Fujisawa T, Ikegami H, Yamato E, Takekawa K, Nakagawa Y, Hamada Y, et al.
A mutation in the glucagon receptor gene (Gly40Ser): heterogeneity in the association
with diabetes mellitus. Diabetologia 1995;38:983–5.
31. Yamada Y, Hayami T, Nakamura K, Kaisaki PJ, Someya Y, Wang CZ, et al. Human
gastric inhibitory polypeptide receptor: cloning of the gene (GIPR) and cDNA. Geno-
mics 1995;29:773–6.
32. Alana I, Hewage CM, Malthouse JP, Parker JC, Gault VA, O’Harte FP. NMR structure
of the glucose-dependent insulinotropic polypeptide fragment, GIP(1–30)amide.
Biochem Biophys Res Commun 2004;325:281–6.
33. Nogueiras R, Tschop M. Biomedicine. Separation of conjoined hormones yields appe-
tite rivals. Science 2005;310:985–6.
34. Egerod KL, Holst B, Petersen PS, Hansen JB, Mulder J, Hokfelt T, et al. GPR39
splice variants versus antisense gene LYPD1: expression and regulation in gastrointestinal
tract, endocrine pancreas, liver, and white adipose tissue. Mol Endocrinol 2007;21:
1685–98.
35. Popovics P, Stewart AJ. GPR39: a Zn(2þ)-activated G protein-coupled receptor that
regulates pancreatic, gastrointestinal and neuronal functions. Cell Mol Life Sci 2011;68:
85–95.
36. Jackson VR, Nothacker HP, Civelli O. GPR39 receptor expression in the mouse brain.
Neuroreport 2006;17:813–6.
37. Stoffel M, Espinosa 3rd R, Le Beau MM, Bell GI. Human glucagon-like peptide-1
receptor gene. Localization to chromosome band 6p21 by fluorescence in situ hybrid-
ization and linkage of a highly polymorphic simple tandem repeat DNA polymorphism
to other markers on chromosome 6. Diabetes 1993;42:1215–8.
38. Tokuyama Y, Matsui K, Egashira T, Nozaki O, Ishizuka T, Kanatsuka A. Five missense
mutations in glucagon-like peptide 1 receptor gene in Japanese population. Diabetes Res
Clin Pract 2004;66:63–9.
39. Tanizawa Y, Riggs AC, Elbein SC, Whelan A, Donis-Keller H, Permutt MA. Human
glucagon-like peptide-1 receptor gene in NIDDM. Identification and use of simple
sequence repeat polymorphisms in genetic analysis. Diabetes 1994;43:752–7.
40. Wildhage I, Trusheim H, Goke B, Lankat-Buttgereit B. Gene expression of the human
glucagon-like peptide-1 receptor is regulated by Sp1 and Sp3. Endocrinology 1999;140:
624–31.
41. Galehshahi FS, Goke B, Lankat-Buttgereit B. A novel silencer element repressing
expression of the GLP-1 receptor gene in fibroblasts and pancreatic A-cells, but not
in pancreatic B- and D-cells. Peptides 2000;21:1169–76.
42. Al-Sabah S, Donnelly D. A model for receptor-peptide binding at the glucagon-like
peptide-1 (GLP-1) receptor through the analysis of truncated ligands and receptors.
Br J Pharmacol 2003;140:339–46.
43. Parthier C, Kleinschmidt M, Neumann P, Rudolph R, Manhart S, Schlenzig D, et al.
Crystal structure of the incretin-bound extracellular domain of a G protein-coupled
receptor. Proc Natl Acad Sci USA 2007;104:13942–7.
104 Jian V. Zhang et al.
Author's personal copy
44. Lopez de Maturana R, Willshaw A, Kuntzsch A, Rudolph R, Donnelly D. The isolated
N-terminal domain of the glucagon-like peptide-1 (GLP-1) receptor binds exendin pep-
tides with much higher affinity than GLP-1. J Biol Chem 2003;278:10195–200.
45. Mann RJ, Al-Sabah S, de Maturana RL, Sinfield JK, Donnelly D. Functional coupling of
Cys-226 and Cys-296 in the glucagon-like peptide-1 (GLP-1) receptor indicates a
disulfide bond that is close to the activation pocket. Peptides 2010;31:2289–93.
46. Dezelak M, Bavec A. Glucagon like-peptide-1 receptor is covalently modified by
endogenous mono-ADP-ribosyltransferase. Mol Biol Rep 2012;39:4375–81.
47. Bullock BP, Heller RS, Habener JF. Tissue distribution of messenger ribonucleic acid
encoding the rat glucagon-like peptide-1 receptor. Endocrinology 1996;137:2968–78.
48. Campos RV, Lee YC, Drucker DJ. Divergent tissue-specific and developmental expres-
sion of receptors for glucagon and glucagon-like peptide-1 in the mouse. Endocrinology
1994;134:2156–64.
49. Storjohann L, Holst B, Schwartz TW. Molecular mechanism of Zn2 þagonism in the
extracellular domain of GPR39. FEBS Lett 2008;582:2583–8.
50. Elrick H, Stimmler L, Hlad Jr CJ, Arai Y. Plasma insulin response to oral and intravenous
glucose administration. J Clin Endocrinol Metab 1964;24:1076–82.
51. Kazakos K. Incretin effect: GLP-1, GIP, DPP4. Diabetes Res Clin Pract 2011;93(Suppl. 1):
S32–S36.
52. Scrocchi LA, Brown TJ, MaClusky N, Brubaker PL, Auerbach AB, Joyner AL, et al.
Glucose intolerance but normal satiety in mice with a null mutation in the glucagon-like
peptide 1 receptor gene. Nat Med 1996;2:1254–8.
53. Montrose-Rafizadeh C, Avdonin P, Garant MJ, Rodgers BD, Kole S, Yang H, et al.
Pancreatic glucagon-like peptide-1 receptor couples to multiple G proteins and activates
mitogen-activated protein kinase pathways in Chinese hamster ovary cells. Endocrinology
1999;140:1132–40.
54. Wheeler MB, Lu M, Dillon JS, Leng XH, Chen C, Boyd 3rd AE. Functional expression
of the rat glucagon-like peptide-I receptor, evidence for coupling to both adenylyl
cyclase and phospholipase-C. Endocrinology 1993;133:57–62.
55. Drucker DJ, Philippe J, Mojsov S, Chick WL, Habener JF. Glucagon-like peptide I
stimulates insulin gene expression and increases cyclic AMP levels in a rat islet cell line.
Proc Natl Acad Sci USA 1987;84:3434–8.
56. Salapatek AM, MacDonald PE, Gaisano HY, Wheeler MB. Mutations to the third
cytoplasmic domain of the glucagon-like peptide 1 (GLP-1) receptor can functionally
uncouple GLP-1-stimulated insulin secretion in HIT-T15 cells. Mol Endocrinol
1999;13:1305–17.
57. Takhar S, Gyomorey S, Su RC, Mathi SK, Li X, Wheeler MB. The third cytoplasmic
domain of the GLP-1[7–36 amide] receptor is required for coupling to the adenylyl
cyclase system. Endocrinology 1996;137:2175–8.
58. Hallbrink M, Holmqvist T, Olsson M, Ostenson CG, Efendic S, Langel U. Different
domains in the third intracellular loop of the GLP-1 receptor are responsible for
Galpha(s) and Galpha(i)/Galpha(o) activation. Biochim Biophys Acta 2001;1546:79–86.
59. Mathi SK, Chan Y, Li X, Wheeler MB. Scanning of the glucagon-like peptide-1 recep-
tor localizes G protein-activating determinants primarily to the N terminus of the third
intracellular loop. Mol Endocrinol 1997;11:424–32.
60. Dun SL, Brailoiu GC, Brailoiu E, Yang J, Chang JK, Dun NJ. Distribution and biolog-
ical activity of obestatin in the rat. J Endocrinol 2006;191:481–9.
61. Zhao CM, Furnes MW, Stenstrom B, Kulseng B, Chen D. Characterization of
obestatin- and ghrelin-producing cells in the gastrointestinal tract and pancreas of rats:
an immunohistochemical and electron-microscopic study. Cell Tissue Res 2008;331:
575–87.
105Obestatin Receptor in Energy Homeostasis and Obesity Pathogenesis
Author's personal copy
62. Gronberg M, Tsolakis AV, Magnusson L, Janson ET, Saras J. Distribution of obestatin
and ghrelin in human tissues: immunoreactive cells in the gastrointestinal tract, pancreas,
and mammary glands. J Histochem Cytochem 2008;56:793–801.
63. Bresciani E, Rapetti D, Dona F, Bulgarelli I, Tamiazzo L, Locatelli V, et al. Obestatin
inhibits feeding but does not modulate GH and corticosterone secretion in the rat.
J Endocrinol Invest 2006;29:RC16–RC18.
64. Green BD, Irwin N, Flatt PR. Direct and indirect effects of obestatin peptides on food
intake and the regulation of glucose homeostasis and insulin secretion in mice. Peptides
2007;28:981–7.
65. Brunetti L, Leone S, Orlando G, Recinella L, Ferrante C, Chiavaroli A, et al. Effects of
obestatin on feeding and body weight after standard or cafeteria diet in the rat. Peptides
2009;30:1323–7.
66. Sibilia V, Bresciani E, Lattuada N, Rapetti D, Locatelli V, De Luca V, et al. Intra-
cerebroventricular acute and chronic administration of obestatin minimally affect food
intake but not weight gain in the rat. J Endocrinol Invest 2006;29:RC31–RC34.
67. Zizzari P, Longchamps R, Epelbaum J, Bluet-Pajot MT. Obestatin partially affects
ghrelin stimulation of food intake and growth hormone secretion in rodents. Endocrinol-
ogy 2007;148:1648–53.
68. Nogueiras R, Pfluger P, Tovar S, Arnold M, Mitchell S, Morris A, et al. Effects of
obestatin on energy balance and growth hormone secretion in rodents. Endocrinology
2007;148:21–6.
69. Seoane LM, Al-Massadi O, Pazos Y, Pagotto U, Casanueva FF. Central obestatin admin-
istration does not modify either spontaneous or ghrelin-induced food intake in rats.
J Endocrinol Invest 2006;29:RC13–RC15.
70. Gourcerol G, Coskun T, Craft LS, Mayer JP, Heiman ML, Wang L, et al. Preproghrelin-
derived peptide, obestatin, fails to influence food intake in lean or obese rodents. Obesity
2007;15:2643–52.
71. Kobelt P, Wisser AS, Stengel A, Goebel M, Bannert N, Gourcerol G, et al. Peripheral
obestatin has no effect on feeding behavior and brain Fos expression in rodents. Peptides
2008;29:1018–27.
72. Gourcerol G, St-Pierre DH, Tache Y. Lack of obestatin effects on food intake: should
obestatin be renamed ghrelin-associated peptide (GAP)? Regul Pept 2007;141:1–7.
73. De Spiegeleer B, Vergote V, Pezeshki A, Peremans K, Burvenich C. Impurity profiling
quality control testing of synthetic peptides using liquid chromatography-photodiode
array-fluorescence and liquid chromatography-electrospray ionization-mass spectrome-
try: the obestatin case. Anal Biochem 2008;376:229–34.
74. Ataka K, Inui A, Asakawa A, Kato I, Fujimiya M. Obestatin inhibits motor activity in the
antrum and duodenum in the fed state of conscious rats. Am J Physiol Gastrointest Liver
Physiol 2008;294:G1210–G1218.
75. Fujimiya M, Asakawa A, Ataka K, Kato I, Inui A. Different effects of ghrelin, des-acyl
ghrelin and obestatin on gastroduodenal motility in conscious rats. World J Gastroenterol
2008;14:6318–26.
76. Depoortere I, Thijs T, Moechars D, De Smet B, Ver Donck L, Peeters TL. Effect of
peripheral obestatin on food intake and gastric emptying in ghrelin-knockout mice.
Br J Pharmacol 2008;153:1550–7.
77. De Smet B, Thijs T, Peeters TL, Depoortere I. Effect of peripheral obestatin on gastric
emptying and intestinal contractility in rodents. Neurogastroenterol Motil 2007;19:211–7.
78. Gurriaran-Rodriguez U, Al-Massadi O, Roca-Rivada A, Crujeiras AB, Gallego R,
Pardo M, et al. Obestatin as a regulator of adipocyte metabolism and adipogenesis. J Cell
Mol Med 2011;15:1927–40.
106 Jian V. Zhang et al.
Author's personal copy
79. Miegueu P, St Pierre D, Broglio F, Cianflone K. Effect of desacyl ghrelin, obestatin and
related peptides on triglyceride storage, metabolism and GHSR signaling in 3T3-L1 ad-
ipocytes. J Cell Biochem 2011;112:704–14.
80. Agnew A, Calderwood D, Chevallier OP, Greer B, Grieve DJ, Green BD. Chronic
treatment with a stable obestatin analog significantly alters plasma triglyceride levels
but fails to influence food intake; fluid intake; body weight; or body composition in rats.
Peptides 2011;32:755–62.
81. Pi-Sunyer FX. The obesity epidemic: pathophysiology and consequences of obesity.
Obes Res 2002;10(Suppl. 2):97S–104S.
82. Guo ZF, Zheng X, Qin YW, Hu JQ, Chen SP, Zhang Z. Circulating preprandial
ghrelin to obestatin ratio is increased in human obesity. J Clin Endocrinol Metab
2007;92:1875–80.
83. Nakahara T, Harada T, Yasuhara D, Shimada N, Amitani H, Sakoguchi T, et al. Plasma
obestatin concentrations are negatively correlated with body mass index, insulin resis-
tance index, and plasma leptin concentrations in obesity and anorexia nervosa. Biol
Psychiatry 2008;64:252–5.
84. Zamrazilova H, Hainer V, Sedlackova D, Papezova H, Kunesova M, Bellisle F, et al.
Plasma obestatin levels in normal weight, obese and anorectic women. Physiol Res
2008;57(Suppl. 1):S49–S55.
85. Lippl F, Erdmann J, Lichter N, Tholl S, Wagenpfeil S, Adam O, et al. Relation of plasma
obestatin levels to bmi, gender, age and insulin. Horm Metab Res 2008;40:806–12.
86. Huda MS, Durham BH, Wong SP, Deepak D, Kerrigan D, McCulloch P, et al. Plasma
obestatin levels are lower in obese and post-gastrectomy subjects, but do not change in
response to a meal. Int J Obes 2008;32:129–35.
87. Vicennati V, Genghini S, De Iasio R, Pasqui F, Pagotto U, Pasquali R. Circulating
obestatin levels and the ghrelin/obestatin ratio in obese women. Eur J Endocrinol
2007;157:295–301.
88. Zou CC, Liang L, Wang CL, Fu JF, Zhao ZY. The change in ghrelin and obestatin levels
in obese children after weight reduction. Acta Paediatr 2009;98:159–65.
89. Zhang N, Yuan C, Li Z, Li J, Li X, Li C, et al. Meta-analysis of the relationship between
obestatin and ghrelin levels and the ghrelin/obestatin ratio with respect to obesity. Am J
Med Sci 2011;341:48–55.
90. Gao XY, Kuang HY, Liu XM, Ma ZB. Decreased gastric body mucosa obestatin expres-
sion in overweight and obese patients. Peptides 2010;31:291–6.
91. Roth CL, Reinehr T, Schernthaner GH, Kopp HP, Kriwanek S, Schernthaner G.
Ghrelin and obestatin levels in severely obese women before and after weight loss after
Roux-en-Y gastric bypass surgery. Obes Surg 2009;19:29–35.
92. Martins C, Kjelstrup L, Mostad IL, Kulseng B. Impact of sustained weight loss achieved
through Roux-en-Y gastric bypass or a lifestyle intervention on ghrelin, obestatin, and
ghrelin/obestatin ratio in morbidly obese patients. Obes Surg 2011;21:751–8.
93. Manshouri M, Ghanbari-Niaki A, Kraemer RR, Shemshaki A. Time course alterations
of plasma obestatin and growth hormone levels in response to short-term anaerobic
exercise training in college women. Appl Physiol Nutr Metab 2008;33:1246–9.
107Obestatin Receptor in Energy Homeostasis and Obesity Pathogenesis
Author's personal copy
... Obestatin is a recently discovered 23-amino acid peptide encoded by the ghrelin gene [155,156] . Although in the original Zhang's works obestatin appeared to suppress food intake and decrease gastric emptying [155][156][157] , consequently antagonizing the orexigenic effect of ghrelin, subsequent researches in rodents showed controversial results [155][156][157][158][159][160][161] . ...
... Obestatin is a recently discovered 23-amino acid peptide encoded by the ghrelin gene [155,156] . Although in the original Zhang's works obestatin appeared to suppress food intake and decrease gastric emptying [155][156][157] , consequently antagonizing the orexigenic effect of ghrelin, subsequent researches in rodents showed controversial results [155][156][157][158][159][160][161] . Obestatin is present not only in the gastrointestinal tract, but also in the spleen, mammary gland, breast milk, and plasma [155,156,160] . ...
... Obestatin is a recently discovered 23-amino acid peptide encoded by the ghrelin gene [155,156] . Although in the original Zhang's works obestatin appeared to suppress food intake and decrease gastric emptying [155][156][157] , consequently antagonizing the orexigenic effect of ghrelin, subsequent researches in rodents showed controversial results [155][156][157][158][159][160][161] . Obestatin is present not only in the gastrointestinal tract, but also in the spleen, mammary gland, breast milk, and plasma [155,156,160] . ...
Article
Full-text available
Obesity and its associated diseases are a worldwide epidemic disease. Usual weight loss cures - as diets, physical activity, behavior therapy and pharmacotherapy - have been continuously implemented but still have relatively poor long-term success and mainly scarce adherence. Bariatric surgery is to date the most effective long term treatment for morbid obesity and it has been proven to reduce obesity-related co-morbidities, among them nonalcoholic fatty liver disease, and mortality. This article summarizes such variations in gut hormones following the current metabolic surgery procedures. The profile of gut hormonal changes after bariatric surgery represents a strategy for the individuation of the most performing surgical procedures to achieve clinical results. About this topic, experts suggest that the individuation of the crosslink among the gut hormones, microbiome, the obesity and the bariatric surgery could lead to new and more specific therapeutic interventions for severe obesity and its co-morbidities, also non surgical.
... Obestatin was initially discovered as the cognate ligand for the orphan G-proteincoupled receptor GPR39 [4,6]. However, these findings were later challenged by several independent groups that were unable to confirm that obestatin has agonist properties on GPR39 [5,7,8]. More recently, obestatin binding to the glucagon-like peptide 1 receptor (GLP-1R) in pancreatic β-cells and adipocytes has been proposed, in line with the effects of GLP-1 and its analogs in these cells [9,10]. ...
... With regard to its central activities, besides counteracting ghrelin effects on food intake and GH secretion, obestatin was found to exert its own effects, by suppressing food intake, slowing gastric emptying and jejunal motility and reducing body weight gain in rodents [4,6,27]. Despite these findings, obestatin central effects are still controversial, as studies in rats and mice also showed no inhibitory effect on food intake and body weight gain, in both the absence or presence of ghrelin [7,8]. In addition, obestatin inhibits thirst and influences memory, anxiety, and sleep likely through indirect action on vagal neurons or by regulating secretion of other hormones that may reach the target cells [28][29][30]. ...
... Interestingly, obestatin binding is displaced by the GLP-1R agonist Ex-4 and by the antagonist Ex-9, and prevented by small interfering RNA (siRNA) against GLP-1R, suggesting possible interaction with GLP-1R [9]. Besides the debated role of GPR39, additional studies in these and other cell types are needed to clarify whether GLP-1R may be or not a putative obestatin receptor [7]. ...
Article
Full-text available
Obestatin was identified in 2005 by Zhang and colleagues as a ghrelin-associated peptide, derived from posttranslational processing of the prepro-ghrelin gene. Initially, obestatin was reported to activate the G-protein-coupled receptor GPR39 and to reduce food intake and gastric emptying. However, obestatin remains a controversial peptide, as these findings have been questioned and its receptor is still a matter of debate, as well as its effects on feeding behavior. Recently, interaction with the glucagon-like peptide 1 receptor has been suggested, in line with obestatin-positive effects on glucose and lipid metabolism. In addition, obestatin displays a variety of cellular effects, by regulating metabolic cell functions, increasing cell survival and proliferation, and inhibiting apoptosis and inflammation in different cell types. Finally, like ghrelin, obestatin is produced in the gastrointestinal tract, including the pancreas and adipose tissue, and exerts both local actions in peripheral tissues, and distant effects at the central level. Therefore, obestatin may indeed be considered a hormone, although additional studies are required to clarify its physiopathological role and to definitely identify its receptor. © 2014 S. Karger AG, Basel.
... The initial hypothesis indicated that obestatin probably binds to the GPR39 receptor or GLP-1R receptor of pancreatic β-cells [18], but studies did not confirm this thesis. So far, the expression of obestatin in the CNS has not been established; however, it is believed that it may participate in the activity of the brain-gut axis [19]. ...
... The potential role of obestatin is associated with chronic metabolic disorders. Numerous studies have demonstrated a negative correlation between obestatin and serum insulin levels [19]. Obestatin also correlates inversely with Body Mass Index (BMI), leptin, glucose levels, and homeostatic model assessment for insulin resistance (HOMA-IR index); thus, it is considered a protein with an essential role in the pathogenesis of diabetes [20,21]. ...
Article
Full-text available
Anorexia nervosa (AN) is an eating disorder characterized by restrictive eating and significant weight loss. In the course of AN, changes are observed in appetite regulation, including orexigenic ghrelin and potentially anorexigenic obestatin. The study aimed to determine if any changes in serum ghrelin and obestatin levels during treatment of AN are observed, while investigating the correlations between these peptides and the severity of disturbed eating attitudes, depression, and anxiety. Thirty adolescent inpatients with AN (examined twice: before hospitalization treatment AN-BT and after treatment AN-AT) and thirty healthy age- and height-matched girls (CG) participated in the study. Anthropometric, serum ghrelin and obestatin concentrations and psychometric evaluations (Eating Attitudes Test 26 Item-EAT-26, Beck Depression Inventory-BDI, Hamilton Depression Rating Scale-HDRS, and Yale Brown Obsessive-Compulsive Scale-Y-BOCS) were performed. The study revealed significantly higher ghrelin and obestatin levels in AN-BT than in AN-AT. A trend toward lower levels during treatment provided partial normalizations. Analyzing correlations in the AN-BT vs. CG group, correlations of peptides with EAT-26, BDI, and HDRS scores were detected. These results suggest a potential role for ghrelin and obestatin in the context of defense mechanisms regulating appetite and body weight in the course of AN and in terms of psychopathological changes co-occurring with this eating disorder.
... Initially obestatin was reported to activate the G-protein-coupled receptor, GPR39 [1,15], but later studies did not confirm that obestatin was a ligand of this receptor [7,[16][17][18][19][20]. More recently Granata et al. described that obestatin may bind to the glucagon-like peptide 1 receptor (GLP-1R) in adipocytes and pancreatic beta cells [8,21]. ...
... Obestatin seemed to present similar effects on the inflammatory processes in the gut [67][68][69][70]74,75]. Our presented study showed that obestatin reduced the severity of TNBS-induced colitis in rats; however, the direct mechanism of its action remains unclear [1,7,8,[15][16][17][18][19][20][21]. ...
Article
Full-text available
Obestatin is a 23-amino acid peptide derived from proghrelin, a common prohormone for ghrelin and obestatin. Previous studies showed that obestatin exhibited some protective and therapeutic effects in the gut. The aim of our presented study was to examine the effect of treatment with obestatin on trinitrobenzene sulfonic acid (TNBS)-induced colitis. In rats anesthetized with ketamine, colitis was induced through intrarectal administration of 25 mg of 2,4,6-trinitrobenzene sulfonic acid (TNBS). Obestatin was administered intraperitoneally at doses of 4, 8, or 16 nmol/kg, twice per day for four consecutive days. The first dose of obestatin was given one day before the induction of colitis, and the last one was given two days after administration of TNBS. Fourteen days after the induction of colitis, rats were anesthetized again with ketamine, and the severity of colitis was determined. The administration of obestatin had no effect on the parameters tested in rats without the induction of colitis. In rats with colitis, administration of obestatin at doses of 8 or 16 nmol/kg reduced the area of colonic damage, and improved mucosal blood flow in the colon. These effects were accompanied by a reduction in the colitis-evoked increase in the level of blood leukocytes, and mucosal concentration of pro-inflammatory interleukin-1β. Moreover, obestatin administered at doses of 8 or 16 nmol/kg reduced histological signs of colonic damage. The administration of obestatin at a dose of 4 nmol/kg failed to significantly affect the parameters tested. Overall, treatment with obestatin reduced the severity of TNBS-induced colitis in rats. This effect was associated with an improvement in mucosal blood flow in the colon, and a decrease in local and systemic inflammatory processes.
... Interestingly, obestatin and ghrelin derive from the same peptide precursor (preprogrelin), and obestatin has opposite effects than ghrelin (15). In studies conducted to date, obestatin has been shown to be particularly effective in disorders of energy homeostasis such as obesity (16,17). In gynecological studies, many different subjects have been studied, but the most marked changes have been observed in PCOS, which is thought to be associated with obesity (18,19). ...
Article
Background: The rising incidence of endometrial cancer has been associated with increases in obesity and physical inactivity. We investigated the clinical significance of serum obestatin levels in women with endometrial cancer, endometrial hyperplasia, and age-matched healthy controls. Material and Methods: The present study was a case-control study conducted at a single center between May 2014 and July 2015. The medical records of 90 patients with a final histopathologic diagnosis after therapeutic curettage for abnormal uterine bleeding were reviewed and categorized by diagnosis. The study population included women with adenocarcinoma of the endometrium (n= 33), hyperplasia of the endometrium (n= 27), and proliferative endometrium (n=30) according to histopathological diagnosis. The women with proliferative endometrium formed the control group. Results: Endometrial cancer was diagnosed in 33 (36.6%) of the patients who presented to our clinic for abnormal uterine bleeding. In the group with endometrial cancer, the mean age was 55.2 ± 8.6 years. There were no differences in obestatin levels between groups (p > 0.05). In the ROC curve analysis, the area under the curve value was 0.574, and obestatin did not prove to be a significant marker for cancer prediction in the population involved in the study. Conclusion: This study did not demonstrate a clear association between circulating levels of obestatin and endometrial pathologies.
... It is a 23-amino acid peptide encoded by the same gene that encodes ghrelin [2]. It appears to function as a part of a complex gut-brain network, whereby hormones and substances from the stomach and intestines signal the brain about satiety and hunger [3]. ...
Article
Full-text available
Aim This work was performed to study the effect of obestatin on normal, diabetic, and obese male albino rats. Materials and methods A total of 60 male albino rats were divided into six groups: group 1 was the normal control group; group 2, the obestatin-treated group, was injected with 100 μg/kg of obestatin subcutaneously twice daily for 1 week; group 3 was the diabetic control group; group 4, the obestatin-treated diabetic group, was injected with obestatin as in group 2; group 5 was the obese control group; and group 6, the obestatin-treated obese group, was injected with obestatin as in group 2. At the end of the experimental period, serum samples were collected after decapitation for estimation of fasting glucose, insulin, triglyceride (TG), and total cholesterol. BMI was also measured. Results After 1 week of obestatin treatment, the obestatin-treated control group showed insignificant changes in serum fasting glucose, insulin, TG, and total cholesterol levels with the exception of BMI, which showed a significant decrease as compared with the normal control group. However, the obestatin-treated diabetic group showed a significant decrease in serum fasting glucose, TG, and total cholesterol levels, with a significant increase in insulin level, but an insignificant change in BMI as compared with the diabetic control group. In contrast, the obestatin-treated obese group showed insignificant changes in serum fasting glucose and insulin, but significant decrease in serum TG, total cholesterol levels, and BMI as compared with the obese control group. Conclusion From these results, we conclude that there is a significant role for obestatin in the regulation of carbohydrate and lipid metabolism, shown from its effect on glucose, insulin, and serum lipids. Thus, obestatin administration develops new and more efficient therapeutic approaches to treat diabetes and obesity.
... Interestingly, obestatin binding was displaced by the GLP-1R agonist Ex-4 and by the antagonist Ex-9, and was prevented by small interfering RNA (siRNA) against GLP-1R, suggesting that similarly to pancreatic β-cells, the peptide may interact with GLP-1R [ 6 ] . Besides the debated role of GPR39, additional studies in these and other cell types, will be addressed to clarify whether obestatin may be or not a GLP-1R ligand [ 51 ] . In adipocytes, obestatin has been recently shown to exert a variety of eff ects, regulating adipocyte function and both lipid and glucose metabolism. ...
... On the other hand, obestatin, originally thought to act through GPR39, 126 is a peptide derived from preproghrelin with some functions on regulating energy balance and gastric emptying. 127 It has an unknown receptor and GPR39 goes back to be an orphan receptor. GRP39 expression is restricted to endocrine and metabolic organs including the pancreas, the liver, the gastrointestinal tract, and white adipose tissue. ...
Article
As critical regulators of almost all physiological processes in the body, G protein-coupled receptors (GPCRs) are important regulators of glucose homeostasis. Some of the newer drugs for treating diabetes mellitus or in development are also targeting GPCRs. This chapter provides a summary of some of the GPCRs that have been shown to be involved in regulating glucose homeostasis.
Article
Since the discovery of adipose tissue as a higly active endocrine tissue, adipokines, peptides produced by adipose tissue and exerting autocrine, paracrine and endocrine function, have gained increasing interest in various obesity-related diseases, including nonalcoholic fatty liver disease (NAFLD). Data regarding the association between NAFLD and circulating leptin and adiponectin levels are generally well documented: leptin levels increase, whereas adiponectin levels decrease, by increasing the severity of NAFLD. Data regarding other adipokines in histologically confirmed NAFLD populations are inconclusive (e.g., resistin, visfatin, retinol-binding protein-4, chemerin) or limited (e.g., adipsin, obestatin, omentin, vaspin etc.). This review summarizes evidence on the association between adipokines and NAFLD. The first part of the review provides general consideration on the interplay between adipokines and NAFLD, and the second part provides evidence on specific adipokines possibly involved in NAFLD pathogenesis. A thorough insight into the pathophysiologic mechanisms linking adipokines with NAFLD may result in the design of studies investigating the combined adipokine use as noninvasive diagnostic markers of NAFLD and new clinical trials targeting the treatment of NAFLD.
Article
Obestatin is a 23 amino acid amidated peptide, member of the preproghrelin gene-derived peptides. Initially, obestatin was reported to exert opposite effects to those of ghrelin on food intake and body weight gain, through interaction with GPR39; however, these findings are still strongly debated and obestatin biological role remains largely unknown. Interestingly, binding of obestatin to the glucagon-like peptide 1 receptor has been recently suggested. Despite being a controversial peptide, recent findings have clearly indicated that obestatin is indeed a multifunctional peptide, exerting a variety of effects, such as stimulation of cell proliferation, survival and differentiation, influence on glucose and lipid metabolism, as well as anti-inflammatory and cardioprotective actions. Its positive effects on glucose and lipid metabolism candidate this peptide as a potential therapeutic tool in pathological conditions such as insulin resistance and diabetes. © 2013 IUBMB Life, 2013.
Article
Full-text available
Chinese hamster ovary (CHO) cells stably expressing the human insulin receptor and the rat glucagon-like peptide-1 (GLP-1) receptor (CHO/GLPR) were used to study the functional coupling of the GLP-1 receptor with G proteins and to examine the regulation of the mito- gen-activated protein (MAP) kinase signaling pathway by GLP-1. We showed that ligand activation of GLP-1 receptor led to increased incorporation of GTP-azidoanilide into Gsa ,G q/11a, and Gi1,2a, but not Gi3a. GLP-1 increased p38 MAP kinase activity 2.5- and 2.0-fold over the basal level in both CHO/GLPR cells and rat insulinoma cells (RIN 1046 -38), respectively. Moreover, GLP-1 induced phosphorylation of the immediate upstream kinases of p38, MKK3/MKK6, in CHO/ GLPR and RIN 1046 -38 cells. Ligand-stimulated GLP-1 receptor produced 1.45- and 2.7-fold increases in tyrosine phosphorylation of 42-kDa extracellular signal-regulated kinase (ERK) in CHO/GLPR and RIN 1046 -38 cells, respectively. In CHO/GLPR cells, these effects of GLP-1 on the ERK and p38 MAP kinase pathways were inhibited by pretreatment with cholera toxin (CTX), but not with pertussis toxin. The combination of insulin and GLP-1 resulted in an additive response (1.6-fold over insulin alone) that was attenuated by CTX. In contrast, the ability of insulin alone to activate these pathways was insensitive to either toxin. Our study indicates a direct coupling be- tween the GLP-1 receptor and several G proteins, and that CTX- sensitive proteins are required for GLP-1-mediated activation of MAP kinases. (Endocrinology 140: 1132-1140, 1999)
Article
Full-text available
The human glucagon-like peptide-1 (GLP-1) receptor mediates the insulinotropic effects of the incretin hormone GLP-1. It is expressed in a cell- and tissue-specific manner. Recently, we cloned the 59-region of the GLP-1 receptor gene and found that tissue and cell specificity is lost by 59-deletion to 2574. In this region proximal to the main transcription start point three putative binding sites for Sp1 were localized. Now, in vitro binding of Sp1 was shown by deoxyribonu- clease footprint analysis with DNA fragments using either recombi- nant Sp1 or nuclear extracts from HIT cells. To elucidate the roles of the three Sp1-binding sites, we mutated each of the sites individually as well as in different combinations. The activity of each construct was analyzed in comparison to the wild-type promoter. Mutation of two adjacent Sp1-binding sites showed a clear reduction of activity. Con- trasting results were obtained after mutation of the third, more distal Sp1-binding site. Here, a clear increase (;150%) revealed a silencing effect of this cis-regulatory element, possibly resembling a Sp3-bind- ing site. Electrophoretic mobility shift analysis revealed binding of Sp1 and Sp3, which was demonstrated by supershifts using specific antibodies. Cotransfection with Sp1 and Sp3 expression vectors in insect cells lacking endogenous Sp factors clearly demonstrated the involvement of Sp1 and Sp3. Therefore, the basal activity of the GLP-1 receptor gene is mediated by two proximal Sp1-binding sites, whereas a more distal site acts as a repressor. (Endocrinology 140: 624 - 631, 1999)
Article
A possible pathogenic mutation in the glucagon receptor gene causing a Gly to Ser change at codon 40 (Gly40Ser) was reported to be associated and linked with non-insulin-dependent diabetes mellitus (NIDDM), in France and Sardinia, Since the frequency of the mutation (Gly40Ser), about 5% in the French population of familial NIDDM and 8% in randomly chosen diabetic patients in Sardinia, was much higher than that of any of the previously reported mutations in candidate genes, it is important to clarify whether the contribution of this mutation to NIDDM is universal. In this study, we investigated the association of this mutation with diabetes mellitus in a large number of Japanese diabetic patients (383 NIDDM and 53 insulin-dependent diabetic patients) by polymerase chain reaction-restriction fragment length polymorphism analysis. None of the Japanese diabetic patients showed Gly40Ser mutation and the association of this mutation with NIDDM was significantly different (p<4·10−5 vs French, p<3·10−6 vs Sardinian by Fisher's exact test). The results not only indicate that the mutation plays little, if any, role in susceptibility to diabetes in Japan, but also indicate the genetic heterogeneity in NIDDM and further emphasize the importance of studies on genetic susceptibility to NIDDM and other complex traits in different ethnic groups.
Article
Obesity has reached epidemic proportions in the United States: more than 20% of adults are clinically obese as defined by a body mass index of 30 kg/m 2 or higher, and an additional 30% are overweight. Environmental, behavioral, and genetic factors have been shown to contribute to the development of obesity. Elevated body mass index, particularly caused by abdominal or upper-body obesity, has been associated with a number of diseases and metabolic abnormalities, many of which have high morbidity and mortality. These include hyperinsulinemia, insulin resistance, type 2 diabetes, hypertension, dyslipidemia, coronary heart disease, gallbladder disease, and certain malignancies. This underscores the importance of identifying people at risk for obesity and its related disease states.
Article
Truncated forms of glucagon-like peptide-1 are the most potent endogenous stimuli of insulin secretion and have powerful antidiabetogenic effects. To determine the structure and coupling mechanisms of the human GLP-1 receptor we have isolated two pancreatic islet cDNAs, encoding the 463 amino acid receptor and differing mainly in their 3' untranslated regions. The deduced amino acid sequence is 90% homologous with the rat GLP-1 receptor. Northern blot analysis shows expression of a single 2.7 kb transcript in pancreatic tissue. When expressed in COS-7 cells the recombinant receptor conferred specific, high affinity GLP-1(7-37) binding. GLP-1(7-37) increased intracellular cAMP in a concentration dependent manner and caused an increase in the free cytosolic calcium ([Ca2+]i) from an intracellular pool, characteristic of phospholipase C (PLC) activation. Thus, like the structurally related glucagon and parathyroid hormone receptors, the human GLP-1 receptor can activate multiple intracellular signaling pathways including adenylyl cyclase and PLC. Knowledge of the GLP-1 receptor structure will facilitate the development of receptor agonists and elucidation of the important role of GLP-1 in normal physiology and disease states.
Article
Truncated forms of glucagon-like peptide-1 (tGLP-1) are potent endogenous stimuli of insulin secretion from pancreatic beta cells and have powerful antidiabetogenic effects. In the present study we sought to determine the precise regions of the tGLP-1 receptor (R) that are required for its efficient coupling to the adenylyl cyclase (AC) system since it is well established that cAMP is the primary second messenger activated by tGLP-1. The predicted third intracellular loop (IC3) of the rat tGLP-1R was systemically scanned using a mutagenic based strategy. The resulting receptor mutants were expressed in COS-7 cells and examined for cAMP formation in response to tGLP-1 stimulation (10nM) and [125I] tGLP-1(7-36) amide binding. A single block deletion (IC3-1) within the N-terminal region of IC3 (K334-L335-K336) resulted in a dramatic reduction in the cAMP response to tGLP-1 (7.1 +/- 1.4% of the wild type (wt) tGLP-1R response, n = 3, p < or = 0.01), while displaying comparable levels of expression, (expressed as the %Bmax of the wt-tGLP-1R (101 +/- 13%, n = 3, p > or = 0.05). This receptor mutation was further analyzed by stable expression in CHO-K1 cells. In agreement with the COS model, IC3-1 displayed comparable levels of receptor expression (97 +/- 16% Bmax of wt tGLP-1R, n = 3, p > or = 0.05) and affinity for tGLP-1(Kd of 460 +/- 15pM vs. 450 +/- 12pM wt tGLP-1R, n = 3, p > or = 0.05), but was unable to effectively stimulate cAMP production (7.7 +/- 0.4% of wt tGLP-1R, n = 3, p < or = 0.01) in response to tGLP-1 (10nM), No other mutation examined within the IC3 domain displayed a lack of correlation between binding activity and cAMP accumulation. Further analysis of the K334-L335-K336 sequence by substitution analysis revealed that a K334 to A substitution was the only modification to result in a striking attenuation of the cAMP response (28 +/- 1.9% of wt tGLP-1, n = 3, p < or = 0.01). These results strongly suggest that within the IC3 domain the N-terminal KLK sequence or a portion thereof (specifically K-334) is required for the efficient coupling of the tGLP-1 receptor to the AC system.
Article
The term incretin effect was used to describe the fact that oral glucose load produces a greater insulin response than that of an isoglycemic intravenous glucose infusion. This difference has been attributed to gastrointestinal peptides GLP-1 and GIP. Since incretin effect is reduced in subjects with type 2 diabetes, despite GLP-1 activity preservation, two forms of incretin-based treatment have emerged: GLP-1R agonists, administered subcutaneously and DPP-4 inhibitors, administered orally. There is a great interest whether incretin-based treatment will be associated with sustained long-term control and improvement in β-cell function. The observation that GLP-1R agonists improve myocardial function and survival of cardiomyocytes highlights the need for further studies. Incretin-based therapies offer a new option and show great promise for the treatment of type 2 diabetes.