ArticlePDF AvailableLiterature Review

GPR120: a critical role in adipogenesis, inflammation, and energy metabolism in adipose tissue

Authors:

Abstract and Figures

It is well known that adipose tissue has a critical role in the development of obesity and metabolic diseases and that adipose tissue acts as an endocrine organ to regulate lipid and glucose metabolism. Accumulating in the adipose tissue, fatty acids serve as a primary source of essential nutrients and act on intracellular and cell surface receptors to regulate biological events. G protein-coupled receptor 120 (GPR120) represents a promising target for the treatment of obesity-related metabolic disorders for its involvement in the regulation of adipogenesis, inflammation, glucose uptake, and insulin resistance. In this review, we summarize recent studies and advances regarding the systemic role of GPR120 in adipose tissue, including both white and brown adipocytes. We offer a new perspective by comparing the different roles in a variety of homeostatic processes from adipogenic development to adipocyte metabolism, and we also discuss the effects of natural and synthetic agonists that may be potential agents for the treatment of metabolic diseases.
Content may be subject to copyright.
Vol.:(0123456789)
1 3
Cell. Mol. Life Sci.
DOI 10.1007/s00018-017-2492-2
REVIEW
GPR120: acritical role inadipogenesis, inflammation, andenergy
metabolism inadipose tissue
TongxingSong1,2· YangYang1,2· YuanfeiZhou1,2· HongkuiWei1,2· JianPeng1,2
Received: 1 November 2016 / Revised: 17 February 2017 / Accepted: 21 February 2017
© Springer International Publishing 2017
Introduction
The global incidence of obesity and type 2 diabetes has
increased over the last three decades. It is well confirmed
that adipose tissues which consist of the white adipose tis-
sues and the brown adipocytes greatly contribute to obe-
sity-associated disease. As the main reservoir to store fatty
acids contain lipid droplets as a type of energy, the white
adipose tissues are to protect and insulate the body at the
physical level [1]. Brown adipocytes, on the other hand,
show high thermogenic capacity and dissipate the stored
energy in the form of heat [2, 3]. Being a remarkably com-
plex organ with significant effects on physiology, adipose
tissue also plays a pivotal role as an endocrine organ in glu-
cose metabolism and immune function through secretion
of a vast range of regulatory factors [4, 5]. Studies on the
developmental and functional roles of adipose tissue have
widely expanded after the 1990s [3]. This paper examines
the functions and regulatory mechanisms of adipose tissue
in the hope of helping to find an efficient way to decrease
the rate of obesity and metabolic diseases.
Free-fatty acids are the basic components of essential
nutrients and serve as signalling molecules that modulate
energy homeostasis [6]. In particular, beneficial effects of
n-3 polyunsaturated fatty acids (n-3 PUFAs) are clearly
observed in anti-inflammatory processes, lipid metabolism,
and glucose homeostasis, as well as in insulin-sensitizing
effects [7, 8]. Notably, the n-3 PUFAs act as ligands of G
protein-coupled receptor 120 (GPR120) [9]. A number of
studies done in the last decade have shown that GPR120
is implicated in crucial homeostatic processes, including
adipogenesis, anti-inflammatory processes, glucose uptake,
and insulin sensitivity, as well as the secretion of hormones,
prompting recent studies to thrust GPR120 into the realm
of drug discovery [1012]. Many excellent recent reviews
Abstract It is well known that adipose tissue has a criti-
cal role in the development of obesity and metabolic dis-
eases and that adipose tissue acts as an endocrine organ to
regulate lipid and glucose metabolism. Accumulating in
the adipose tissue, fatty acids serve as a primary source of
essential nutrients and act on intracellular and cell surface
receptors to regulate biological events. G protein-coupled
receptor 120 (GPR120) represents a promising target for
the treatment of obesity-related metabolic disorders for its
involvement in the regulation of adipogenesis, inflamma-
tion, glucose uptake, and insulin resistance. In this review,
we summarize recent studies and advances regarding the
systemic role of GPR120 in adipose tissue, including both
white and brown adipocytes. We offer a new perspective
by comparing the different roles in a variety of homeo-
static processes from adipogenic development to adipocyte
metabolism, and we also discuss the effects of natural and
synthetic agonists that may be potential agents for the treat-
ment of metabolic diseases.
Keywords GPR120· Fatty acids· White adipose tissue·
Brown adipose tissue· Adipogenesis· Glucose uptake·
Metabolic homeostasis
Cellular and Molecular LifeSciences
* Hongkui Wei
weihongkui@mail.hzau.edu.cn
* Jian Peng
pengjian@mail.hzau.edu.cn
1 Department ofAnimal Nutrition andFeed Science, College
ofAnimal Science andTechnology, Huazhong Agricultural
University, Wuhan430070, China
2 The Cooperative Innovation Center forSustainable Pig
Production, Wuhan430070, China
T.Song et al.
1 3
have provided insights into the biological and physiologi-
cal roles of GPR120 [1316]. However, the detailed role of
GPR120 in adipose tissue has not been well demonstrated
yet.
Hence, the primary goal of this review is to summarize
the most recent and seminal studies that have contributed
to our understanding of the function and regulation of
GPR120, especially in adipose tissue. We first describe the
biological characteristics of GPR120. We then expound on
the inconsistencies and certainties that have been presented
due to advances in our knowledge of the function of adi-
pose tissue. Finally, we discuss the natural and synthetic
agonists that are known to regulate the activity of GPR120
to resist obesity and diabetes.
Biological characteristics ofGPR120
The novel fatty acid receptor GPR120, also known as free-
fatty acid receptor 4 (FFAR4), is a seven transmembrane
receptor and was deorphanised in 2005 when long chain
fatty acids were shown to respond well to it [9, 17]. Numer-
ous studies have reported the characteristics of GPR120
from specific species [1820]. The human GPR120 exists
as two splice variants, and the short variant has a structure
similar to that of the rodent and monkey receptor. Our pre-
vious work reported the three alternatively spliced tran-
scripts of pigs, and the wild-type variant is well matched
with the human short isoform [21]. As a GPCR, GPR120
shares the ability to activate heterotrimeric G proteins and
induces downstream second messenger pathways in the
context of ligand treatment. Hirasawa et al. reported that
the activation of GPR120 by fatty acids augment intracel-
lular calcium (Ca2+) and extracellular signal-regulated
kinase 1/2 (ERK1/2) without affecting the level of cyclic
AMP [17]. Furthermore, Oh et al. showed that GPR120
can effectively respond to n-3 fatty acids by activating
Gαq/11 in adipocytes and by recruiting cytosolic β-arrestin
2 to the cell membrane in macrophages [11]. Hence, we
infer that the various events induced by the two pathways
may involve one of the two coupled proteins, Gαq/11 and
β-arrestin 2. However, other biological roles of GPR120
remain to be explored.
GPR120 has been shown in recent studies to be impli-
cated in diverse physiological homeostasis processes,
including release of gut peptides, insulin sensitization, anti-
inflammatory processes, and regulation of appetite [11, 17,
22]. Consistent with the pleiotropic functions in biological
events, stronger expression of GPR120 is observed in tis-
sues, such as the small intestine, spleen, adipose tissue, and
taste buds. Despite the differing expression pattern from
varied species, a similarly high level of GPR120 is found to
be present in the same tissues, such as the small intestine,
which may reflect the concordant functions, for example,
glucagon-like peptide 1 (GLP-1) secretion [17]. It is worth
noting that GPR120 is found to be expressed endogenously
both in adipocytes and in adipose tissue but not in preadi-
pocytes [12], implying that the role of GPR120 may closely
be related to the development and metabolism of adipose
tissue. This special distribution might reflect the crucial
functions of GPR120.
Physiological functions ofGPR120 inadipose
tissue
The role ofGPR120 inadipose development
The expression ofGPR120 inadipose tissue
A growing number of evidences point to the key role of
GPR120 in adipose development, including both white and
brown adipocytes. As mentioned above, GPR120 cannot be
detected in preadipocytes, showing abundant expression in
mature 3T3-L1 cells (a recognized cell line for white adi-
pocyte research [23]) and human white adipocytes. The
expression of GPR120 increases with MDI treatment (the
differentiation medium containing isobutylmethylxanthine,
dexamethasone, and insulin), a typical adipogenic induc-
tion cocktail for white adipocytes in vitro [23], indicat-
ing that this receptor is probably implicated in adipogenic
differentiation. Using siRNA, Gotoh and his colleagues
reported that GPR120 knockdown inhibited adipogenesis
[12]. Moreover, the adipogenesis process is suppressed in
GPR120−/− mouse embryonic fibroblast-derived (MEF)
adipocytes [10]. Our study also showed that the adipogenic
ability was significantly inhibited in lentivirus-mediated
shGPR120 transfected 3T3-L1 cells [24]. Therefore, we
assume that GPR120 might act as an adipogenic receptor
[25].
An earlier study demonstrated that high expression
of GPR120 was detected in four types of adipose tissues,
including subcutaneous, perinephric, mesenteric, and
epididymis tissue collected from high-fat diet-fed mice
[12]. Experiments in humans showed a similar pattern
revealing a significantly higher expression of GPR120 in
obese individuals than in lean individuals [10]. Another
report, likewise, exhibited a high expression of GPR120 in
the brown adipose tissue (BAT) and the inducement of pro-
tein level in the BAT after cold exposure. It has also been
reported that the activation of GPR120 can promote the
browning of white fat in mice [26]. Thus, it is safe to spec-
ulate that the expression of the receptor may be induced
by high lipid intake in rodents and humans. Although the
exhaustive role of GPR120 is still unclear, its indispensable
function in adipogenesis is fairly certain.
GPR120: acritical role inadipogenesis, inflammation, andenergy metabolism inadipose tissue
1 3
The effects ofGPR120 agonists inadipogenesis
As is well known, with the activation of ligand, the GPCR
couples with G proteins and β-arrestins to transduce a sig-
nal through a downstream pathway, regulating physiologi-
cal events. GPR120 showed a strong response to medium
and long chain polyunsaturated fatty acids (LCPUFA),
especially n-3 PUFAs. Our previous studies showed that
α-linolenic acids (ALA), rather than docosahexaenoic acid
(DHA), improved adipogenesis in a GPR120-dependent
pathway in 3T3L1 cells (Fig.1) [24]. However, when using
a luciferase reporter assay, we found that both DHA and
ALA can effectively activate GPR120 [21]. Despite the
finding that LCPUFA act as natural ligands of GPR120, the
effects of LCPUFA on modulation of 3T3-L1 adipogenic
differentiation are inconclusive.
DHA, similar to other fatty acids, has multifunctional
roles: it may accelerate, inhibit, or do not influence adipo-
genic processes. However, ALA was found to be more sen-
sitive to this receptor in adipogenesis as well as other events
[17]. Therefore, we chose the selective synthetic agonist
TUG-891 to clarify the downstream signalling mediated by
GPR120 and, to some extent, avoiding potential GPR120-
independent mechanisms [27]. Our previous study showed
that pig GPR120 was well activated by TUG-891, and the
stromal vascular fraction cells from porcine adipose tissue,
as well as 3T3-L1 preadipocytes, were promoted through
adipogenesis [24]. Gao etal. reported that TUG-891, at a
low concentration range, functions as a positive enhancer
in the adipogenic process, and higher concentrations induce
the opposite developmental direction, osteogenesis [28].
The different cell types and characteristics and the distinct
expression of GPR120 in the two cells may be the cause of
the diverse mechanisms. On the other hand, GPR120 which
can be definitely stimulated by GW9508, another selective
agonist for GPR120 may induce BAT activity as well as the
n-3 PUFAs [26, 29]. Both the eicosapentaenoic acid (EPA)
and GW9508 were found to have accelerated the adipo-
genesis of brown and beige adipocyte and upregulated
the expression pf key thermogenic genes (Fig.2). On the
whole, GPR120 activation may induce adipogenesis in two
different types of adipocytes, which implies the distinct
roles of this receptor in the two biological events.
What are thecellular effectors ofGPR120 inadipogenesis
During differentiation of 3T3-L1 cells, mRNA expres-
sion of GPR120 closely coincides with the master adipo-
genic regulator, peroxisome proliferator-activated receptor
γ (PPARγ). Interestingly, GPR120 knockdown was found
to reduce the mRNA level of PPARγ and the adipogenic
marker gene, fatty acid binding protein 4 (FABP4) [12].
Moreover, adipogenesis-related genes, including PPARγ
and FABP4, are decreased in adipocytes induced from
GPR120−/− MEF cells [10]. On the other hand, it has been
demonstrated that the PPARγ agonist troglitazone increased
the mRNA expression of GPR120 [12]. These results may
imply that PPARγ is regulated by GPR120 or that GPR120
can interact with PPARγ during adipocyte differentiation.
How is the signal transduced from the membrane recep-
tor to the nuclear receptor? It should be noted that both
GPR120 and PPARγ have similar ligand binding pock-
ets and both bind with DHA [3032]. DHA and ALA are
shown as PPARγ ligands that activate the receptor. In addi-
tion, they can also combine with GPR120 [24]. Therefore,
a selective agonist is necessary for activation of PPARγ.
Although TUG-891 is a potent agonist for pig and mouse
GPR120, less evidence is available regarding the function
of TUG-891 on PPARγ. Using a luciferase reporter system,
Song et al. reported that TUG-891 could activate PPARγ
in a GPR120-dependent manner [24]. This illustrates that
with TUG-891 treatment, activated GPR120 may transduce
the intracellular signal into the nucleus to activate PPARγ.
We speculate that the GPR120-PPARγ pathway may be one
of the main regulation mechanisms to modulate adipogen-
esis in 3T3L1 cells (as shown is Fig. 1). However, using
GW9662 (an inhibitor of PPARγ) in differentiating brown
adipocyte did not dramatically alter the effects of EPA or
Fig. 1 Schematic diagram
of adipogenesis mediated by
GPR120 activation in 3T3L1
cells. With the treatment of n-3
PUFAs or GPR120 synthetic
agonist, TUG-891, GPR120
promotes adipogenesis by acti-
vating PPARγ and elevating the
expression of key adipogenic
gene via [Ca2+]i and ERK1/2
signal pathway in 3T3L1 cells
T.Song et al.
1 3
GW9508 on the phenotype of differentiation, indicating
that PPARγ may be not essential for the EPA- or GW9508-
induced brown adipocyte differentiation in some extent,
and other key downstream regulators, such as uncoupling
protein-1 (UCP1), may play a role consistent with GPR120
in this event [26, 29]. All these suggest that the targets of
the white and brown fat adipogenesis induced by GPR120
are quite different and need to be well examined further.
The downstream signals mediated byGPR120
inadipogenesis
Moreover, the precise signalling mediated by GPR120 is
also worth studying. Numerous studies have shown that
GPR120 coupled with Gαq/11 induces an increase in
intracellular calcium concentration [Ca2+]i and phospho-
rylation of the ERK1/2 cascade [17, 27]. Several studies
have reported that both ERK1/2 and [Ca2+]i play a critical
role in adipogenesis. ERK1/2 facilitates the early stage of
adipogenesis, while this signal needs to be closed at later
stages, which suggests a multifunctional role for ERK1/2
[33, 34]. In addition, there is evidence suggesting that the
[Ca2+]i signal manifests to exert a biphasic function in adi-
pocyte differentiation both in mice and human preadipo-
cytes [35, 36]. In the first 2days of adipogenesis, increas-
ing [Ca2+]i levels suppress this progress but accelerate the
later maturation stage of adipogenic differentiation [35,
36]. The intracellular signal in adipogenesis mediated by
GPR120 remains largely unknown. As shown in Fig. 1,
[Ca2+]i and ERK1/2 may provide new insights to determine
the mechanism of GPR120 in adipogenesis. In 3T3-L1
preadipocytes, TUG-891 activates both of the signals. In
addition, combined treatment with TUG-891 and BAPTA-
AM (a Ca2+ chelator) or U0126 (ERK1/2 inhibitor) abol-
ished the adipogenesis process. Furthermore, PPARγ
mRNA and protein levels are also influenced by blocking
the two signals. This demonstrates that the [Ca2+]i-ERK1/2
pathway is involved in GPR120-induced processes and ulti-
mately targets PPARγ to affect adipogenesis [24]. This may
shed light on one of the possible intracellular pathways. In
BMMSCs, Gao etal. reported that 3days of treatment with
a low concentration of TUG-891 (0.5µM) promoted adipo-
genesis, shown both by the adipogenic phenotype and the
expression of PPARγ [28]. However, the ERK1/2 signal is
blocked in these events. Therefore, the precise underlying
mechanism may be different depending on the cell types
and treatment methods. Others have shown that phospho-
rylation of AKT (p-AKT) cannot be detected in white adi-
pose tissue separated from GPR120−/− mice [10]. Perhaps
p-AKT is also related to GPR120-mediated adipogenesis,
but this association has not been identified. Overall, the
question is how these mentioned signals can be transduced
Fig. 2 Schematic diagram of the role of GPR120 activation in adi-
pose tissues related to the metabolic homeostasis. With the treatment
of n-3 PUFAs or GPR120 synthetic agonists, GPR120 promotes adi-
pogenesis in both white and brown preadipocytes. In white adipose
tissue, GPR120 mediates anti-inflammation and insulin sensitization
effects and GPR120 activation also induces the browning of white
adipocytes. The pleiotropic functions of GPR120 in adipose tissue
will contribute to the whole-body metabolic homeostasis
GPR120: acritical role inadipogenesis, inflammation, andenergy metabolism inadipose tissue
1 3
into nuclear signals to activate PPARγ, and the answer
still needs to be clarified. Given the relationship between
GPR120 and PPARγ in adipogenesis, it will be interest-
ing to demonstrate whether the GPR120-PPARγ pathway
functions in white adipose metabolism and even in other
tissues. In comparison, the down signaling modulated by
GPR120 is somewhat complicated in the brown and beige
adipocytes. It seems that GPR120 activation upregulates
the microRNA 30b/microRNA 378 and the cyclic AMP
(cAMP), in turn, and both microRNAs and cAMP can
affect the level of GPR120 [26, 29].
Considering the especially high expression of GPR120
in adipocytes and adipose tissue, the precise mechanism of
GPR120 in both WAT and BAT is being explored. It is rea-
sonable to say that the role of GPR120 in adipogenesis is
clear, while the important functions in brown/beige adipo-
cytes adipogenesis still need to be clarified.
The metabolic regulation ofGPR120 inadipose tissue
Anti‑inflammatory effects
Obesity gives rise to chronic low-grade inflammation,
which can trigger insulin resistance and type 2 diabetes
[37]. White adipose tissue acts as an important endocrine
organ and releases numerous adipokines that can contrib-
ute to pro- or anti-inflammation states [38]. Adipose tissue
inflammation is a process characterized by an inflammatory
response mediated by adipose tissue macrophages [39].
There is increasing interest focused on the role of adipose
tissue macrophages (ATMs) in obesity and in ATM func-
tions in the inflammatory pathways activated in adipose
tissue, which change the characteristics of obesity in obese
individuals [40, 41]. Moreover, ATMs are highly responsi-
ble for the expression of tumour necrosis factor α (TNFα)
and interleukin-6 (IL-6), which can block normal activity
in adipocytes [42, 43]. Thus, ATMs provide a potential and
pivotal function in the metabolic role of adipose tissue.
Oh etal. analysed the ATMs from wild-type (WT) and
GPR120-deficient mice invivo. The HFD induced the total
number of F4/80 marked M1 macrophages separated from
adipose tissues [11]. In context of n-3 PUFA treatment, the
number of M1 macrophages decreased, while the number
of macrophage galactose-type C-type lectin 1 (MGL1)
marked M2 macrophages strikingly increased in the WT
mice but not in the GPR120-deficient mice [11]. Moreover,
GPR120 also accelerated the M2 macrophage polarization,
which facilitated the formation of an anti-inflammatory sta-
tus in adipose tissue [11]. In addition, adding n-3 PUFA to
the normal diet resulted in decreased macrophage chemo-
taxis capacity in a GPR120-dependent manner. Further-
more, using the synthetic GPR120 agonist cpdA, the posi-
tive effect of GPR120 in the ATMs was confirmed [44].
In view of the above studies, GPR120 functions as an n-3
PUFA sensor in adipose tissue that triggers beneficial anti-
inflammatory effects. In an invitro model, the mechanism
of the anti-inflammatory role has also been elucidated in
RAW 264.7 cells (a type of mouse macrophage). Ligand-
activated GPR120 binds to β-arrestin2, and the complex
interacts with transforming growth factor-β-activated
kinase binding protein 1 (TAB1), in turn, blocking down-
stream key pro-inflammatory signalling molecules, includ-
ing nuclear factor-κB (NFκB) and c-Jun N-terminal kinase
(JNK), and inhibiting inflammation [11]. Other studies
have shown different GPR120-dependent pathways that are
involved with cyclooxygenase 2 (COX-2) expression and
prostaglandin E2 (PGE2) synthesis [45, 46]. These data are
referred in another excellent review [13].
Some metabolins may influence the GPR120-dependent
β-arrestin2 involved in anti-inflammation. It is worth men-
tioning that hyperhomocysteinaemia (HHcy) inhibits insu-
lin sensitivity in adipose tissue and is considered a chronic
inflammatory state by the epidemiological literature [47].
HHcy, characterized by an abnormally high level of homo-
cysteine, acts as a pro-inflammatory factor to induce the
expression and secretion of resistin [48]. Recently, it has
been reported that in rat adipocytes, GPR120 activation
by GW9508 can reverse HHcy-induced insulin resistance
by inhibiting adipose inflammation rather than endoplas-
mic reticulum (ER) stress [49]. GPR120 prevents HHcy-
induced JNK activation and inflammation. Meanwhile, the
downstream signal β-arrestin2 mediated by GPR120 can be
repressed by homocysteine [50]. Thus, it may be interest-
ing to clarify the precise mechanism of the expression and
activation of GPR120-dependent β-arrestin2 involved in
anti-inflammation.
Moreover, it cannot be ignored that the WAT depots,
in the form of triglycerides (TG), store fatty acids derived
from diets, and endogenous synthesis [51]. The lipoly-
sis of TG provides the serum with varying chains FFAs
to influence the macrophage activity [52]. Several excel-
lent reviews focus on the variety of FFAs which modify
the macrophages and other inflammatory cells [5254].
Recently, Rodriguez-Pacheco et al. have reported that
FFAs have different effects on the inflammatory profile
of the adipocytes from the visceral adipocytes tissue and
the pro-inflammatory effects of different fatty acids in the
GPR120 knockdown visceral adipocytes can be weakened
[55]. This is because GPR120 act partially as an inflamma-
tory mediator to regulate the inflammatory effects induced
by the FFAs [55]. In addition, different fatty acids show the
varying degrees of the promotion function on the expres-
sion of GPR120 in both nonobese and morbidly obese
subjects [56]. Perhaps, GPR120 in adipocytes tissues from
different location shows various roles which are based on
the character of the native adipocytes [57]. The observation
T.Song et al.
1 3
of Rodriguez-Pacheco etal. may need to be well dissected
further, especially invivo test [55].
In addition, during adipose tissue remodelling in obesity,
another key factor that we focus on is the role of hypoxia.
Although adipose tissue can recruit new blood vessels dur-
ing expansion, even elevating the oxygen tension in the fat
pads, hypoxia may develop [58]. Moreover, hypoxia-induc-
ible factor 1 (HIF-1) becomes activated in obese adipocytes
[59]. Vascular endothelial grow factor-A (VEGF-A) plays
a critical role in promoting local vascular development in
the growing adipose tissue [60]. Hansan et al. reported
that EPA could increase the release of VEGF-A through
GPR120-dependent and independent pathways in 3T3-
L1 adipocytes [61]. This suggests that the activation of
GPR120 could act in another role to partly promote angi-
ogenesis, which may have an anti-inflammatory func-
tion in adipose tissue [60]. Indeed, given the functions in
both adipocytes and macrophages in the adipose tissue, it
seems possible that GPR120 can act in an anti-diabetic role
through an anti-inflammatory effect in adipose tissue [11,
60]. Currently, several precise mechanisms regarding the
anti-inflammation function of GPR120 in adipose tissue are
being dissected, as well as the interactions among different
cell types [3].
Glucose uptake andenergy metabolism
Consistent with the basic function of fatty acids in energy
metabolism in adipose tissue, the fatty acids receptor,
GPR120, seems to have a pivotal role in regulating adi-
pose metabolism. As described before, GPR120 is found
to be highly expressed in adipocytes and adipose tissue.
With a high-fat diet (HFD) treatment, GPR120-deficient
mice show a notable decrease in adipogenic and lipo-
genic genes, while these genes increased in the liver [10].
Seminal work by Oh etal. has shown that GPR120 acti-
vated by n-3 PUFA and GW9508 leads to insulin sensiti-
zation in vivo and strongly alleviates glucose intolerance
in HFD-induced obesity [11]. In both 3T3-L1 adipocytes
and primary adipose tissue, activated GPR120 signifi-
cantly enhances the activation of the PI3K/Akt pathway,
triggering GLUT4 translocation to the cell membrane and
increasing glucose uptake in a manner dependent on cou-
pling to Gαq/11 not β-arrestin [11]. It is worth mentioning
that Gαq/11 and IRS1 are independent signals that activate
the PI3K-Akt-GLUT4 pathway [11]. One group also devel-
oped a type of GPR120 agonist, compound A (cpdA), that
responded well to insulin sensitivity and improved glucose
tolerance and decreased hyperinsulinemia [44]. Another
small molecule agonist, TUG-891, and members of the
new fatty acid ester of hydroxy fatty acid (9-PAHSA) lipid
class have been designed to activate GPR120 and enhance
glucose uptake [27, 62, 63]. Intriguingly, Liu etal. reported
that in GPR120 knockdown 3T3-L1 cells, the expression of
GLUT4 and insulin receptor substrate 1(IRS1) is decreased
[64]. Ichimura et al. found that insulin signalling-related
genes are remarkably decreased in adipose tissue of HFD-
fed GPR120 −/− mice, especially IRS1 [10]. When the IRS1
level was lower than a specific amount, insulin signalling
was abolished. Therefore, based on the abnormal insulin
signal in adipose tissue and the liver, the GPR120-deficient
mice fed with an HFD diet develop hyperglycaemia, glu-
cose intolerance, and insulin resistance when compared
with the control group. This suggests that dysfunction
of GPR120 may impair normal insulin signalling and
adversely impact glucose uptake in adipose tissue [16].
On the other hand, when mice are challenged by an
HFD, the expression of GPR120 increased higher than
reported in the previous work [12]. Some researchers,
such as Chen et al., have reported an upstream mecha-
nism to regulate GPR120 [65]. In context of the HFD, the
mRNA and protein expression of GPR120 and PPARγ
was increased and the binding of CCAAT/enhancer bind-
ing protein β (C/EBPβ) on the GPR120 core promoter was
accelerated [65]. They speculate that the C/EBPβ-GPR120-
PPARγ pathway might provide a way to regulate energy
metabolism [65]. In addition, another group reported that
EPA upregulated VEGF-A through GPR120-PPARγ or in
a GPR120-independent manner in 3T3-L1 adipocytes [61].
Overexpression of VEGF-A in adipose tissue ameliorated
glucose intolerance and showed an insulin-sensitizing
effect under the challenge of HFD in mice [60]. Thus, this
may demonstrate that in the HFD diet treatment, expres-
sion of GPR120 will be upregulated and activated GPR120
induced by an agonist will improve the metabolic status of
adipose tissue. However, the interaction between GPR120
and PPARγ implicated in energy metabolism needs to be
addressed in the white adipocyte.
Unlike the white adipocytes, the brown and beige adi-
pocytes transfer stored chemical energy in the form of heat
in response to cold or overfeeding [3]. Thus, we can con-
firm that the brown adipocytes thermogenic activity and
the browning of white adipocytes are the key and promis-
ing mechanism to protect body from obesity and metabolic
diseases. Notably, the BAT also contributes as an endocrine
organ [66]. As recently reported, GPR120 may be the cru-
cial mediator in the energy metabolism of brown adipo-
cytes, since n-3 PUFAs- and synthetic agonists-mediated
GPR120 activation induce the release of the hormonal fac-
tor fibroblast growth factor-21 (FGF21), resulting in the
promotion of BAT activity and WAT browning [26].
These data indicate that GPR120 plays a role as a lipid
sensor invivo and senses the dietary fat to regulate glucose
uptake and energy metabolism in adipose tissue, thus trig-
gering a novel pathway of BAT activation and browning of
WAT.
GPR120: acritical role inadipogenesis, inflammation, andenergy metabolism inadipose tissue
1 3
Taken together, the role of GPR120 in anti-inflamma-
tion, glucose uptake, energy metabolism, and other related
biological events, such as angiogenesis, has so far seemed
clear in some extent, although the precise mechanism of
GPR120 needs to be further explored.
What are theinconsistencies duringadipogenesis
andadipocyte metabolism?
The normal expression ofGPR120 may contribute
tothenormal functions
Several studies have reported that GPR120 is required for
normal adipogenesis both in vivo and in vitro. As Gotoh
et al. has shown, GPR120 was increased in four different
adipose tissues from HFD-fed mice [12]. In addition, the
expression of GPR120 was notably increased in subcutane-
ous and visceral adipose tissue from obese individuals [10].
In contrast, another group reported that morbidly obese
individuals have lower mRNA and protein expression of
GPR120 in visceral adipose tissue than lean humans, and
the expression was reduced 3 h after a high-fat meal in
obese people [56]. This is the first inconsistency between
studies. As described above, GPR120 plays an important
role in systemic homeostasis. In GPR120−/− mice, it is easy
to develop obesity, but decreased adipocyte differentiation
with lower PPARγ level and lower content of triglyceride
also occurs [10]. That means the adipogenesis process in
GPR120-deficient mice is abnormal, which is consistent
with the vitro experiment from the Gotoh group [10, 12].
Furthermore, hepatic lipogenesis is higher than in wild-type
mice, which accounts for the final fat and other obesity-
related metabolic problems of GPR120−/− mice. In addi-
tion, it has been indicated that adipogenesis fails to store
all the excess nutrients, and the extra energy will be ectopi-
cally deposited into liver and muscle tissue [67]. Dysfunc-
tion of GPR120 may lead to lipid accumulation in the liver,
resulting in worse insulin resistance [11]. Morbidly obese
individuals may have an absence of functional GPR120 and
the high-fat meal treatment may worsen the dysfunction of
GPR120 and the insulin resistance. If so, the activation of
normal GPR120 can be a way to facilitate positive adipo-
genesis and may improve insulin resistance. However, a
direct method to measure the normal, functional GPR120
in obese people is still missing. Perhaps, the expression of
GPR120 acts as an indicator to assess the state of meta-
bolic homeostasis. Actually, mature adipocytes induce the
production of C16:1n7 palmitoleate, which has been pro-
posed to be a lipid hormone that modulates the interaction
among adipose, muscle, and liver tissue [68]. It seems that
dysfunction of GPR120 leads to insufficient production of
palmitoleate [11, 68].
The contrariety between“the pro‑adipogenic function”
and“anti‑inflammatory role”?
Another inconsistency is between the promoting function in
adipogenesis and the anti-inflammatory role in adipocytes.
The idea that obesity is involved in adipocyte hyperplasia
has created a false notion in our mind: adipogenesis causes
obesity [3]. However, it is worth noting that adipogenesis
occurs in the obesity process but is not the primary driver.
The root cause is the energy balance equation. Given extra
nutrition, increased adipogenesis is driven by a require-
ment to store excess calories. Moreover, the formation of
new adipocytes should be adapted to dispose of the surplus
energy safely [3]. As mentioned before, normal GPR120
may be activated by FFAs to facilitate adipogenesis and
lipogenesis in adipose tissue, thereby resulting in healthy
metabolic homeostasis. Recently, a group of people called
“metabolically healthy obese” (MHO) were identified,
who tend to show reduced visceral adiposity and inflam-
mation and improved glucose and lipid homeostasis [69].
Notably, they have smaller adipocytes than other “normal
obese” people [70]. Another alternative hypothesis is that
adipogenesis increases the number of new, smaller adipo-
cytes that have functional glucose uptake and healthy adi-
pokines to improve metabolic health [3]. Importantly, the
agonist of PPARγ, thiazolidinedione (TZD), plays a pivotal
role in regulating homeostasis and improving insulin resist-
ance. Consequently, TZD increased adipocyte cell numbers
and total adiposity [71], and unhealthy obese people had
a diminished number of preadipocytes [72]. In our previ-
ous work, we found that the expression of GPR120, syn-
chronously with PPARγ, is decreased after 6days of MDI
adipogenic induction [21]. In regard to the hypertrophy
of mature adipocytes, the bigger the adipocyte grows, the
worse their metabolic status. Thus, we speculate that the
increase of GPR120 in adipogenesis may be used for form-
ing new adipocytes, and the decrease of GPR120 in mature
adipocytes may be a response to the dysfunction of adipo-
cytes. However, it seems that making obese people more fat
with new, smaller adipocytes is not a priority for medical
research [3].
Despite some of the vital functions have been revealed,
new roles are still being discovered. The contradictory
nature of the different functions of GPR120 implies that
there is much that is not known about this receptor. The
precise underlying mechanisms of GPR120 signalling need
to be explored. On the whole, it is certain that GPR120 is
required for adipogenesis to format new adipocytes and
functions in metabolic homeostasis via its important, but
not detailed, role in maintaining normal adipose metabo-
lism. Recently, the observation that GPR120 is prominently
increased in murine brown adipose tissue in response to
exposure to cold supports its role in energy expenditure
T.Song et al.
1 3
[73]. Studies have shown that the GPR120 activated by
agonists can trigger the BAT activation and browning of
WAT [ 26, 29]. These findings indicate the importance of
GPR120 in systemic metabolic and further stir the interest
in the potential natural or synthetic agonists of GPR120 for
fighting the metabolic diseases.
The application ofGPR120 agonists inanti‑obesity
andanti‑diabetic therapeutics
As mentioned before, GPR120 is a promising pharmaceu-
tical target for anti-diabetic and anti-obesity treatments.
Hirasawa and colleagues, for the first time, deorphanised
this receptor and found that the ligand was LCFA [17].
After that, others have also identified several types of fatty
acids and have determined that n-3 PUFA is not a unique
ligand for GPR120 [11, 74]. Based on an ectopic GPR120-
overexpression system in HEK293T cells, saturated and
monounsaturated fatty acids have also been found to acti-
vate this receptor [11, 20, 75]. It needs careful considera-
tion regarding the special kind of natural agonists. In some
cell types that have high expression of GPR120, only LCP-
UFA responded well to GPR120. In mouse STC-1 cells,
LCPUFA, but not saturated LCFA, could evoke a clear
response to the secretion of glucagon-like peptide 1(GLP-1)
[17]. Surprisingly, both n-3 and n-6 PUFAs could strongly
activate GPR120 with different signalling events in human
CaCo-2 cells, which are used as an intestinal epithelial
model [76]. Thus, the conclusions should be treated with
caution. In lipidomic analysis experiments, Yore etal. iden-
tified a new class of endogenous agonist, called 9-PAHSA,
and found that it can activate GPR120 in a concentration-
dependent manner [62]. Although 9-PAHSA shares similar
properties with DHA and another agonist, GW9508, the
pharmacological characteristics of this endogenous agonist
have not been well illustrated.
Based on the uncertainty of the natural ligand, it is nec-
essary to explore synthetic ligands with high potency and
selectivity for GPR120. Several recent reviews have well
summarized the development progress of GPR120 ago-
nists. For instance, Briscoe et al. described GW9508, a
small molecule for both GPR120 and GPR40 [74]. In cells
expressing GPR120, but not GPR40, GW9508 can be a
specific agonist for GPR120 [11, 77]. NCG21, another ago-
nist product, was not widely used in subsequent research,
and notably, it is identified among the derivatives of PPARγ
agonists [31]. This may also show the relationship between
GPR120 and PPARγ. Noticeably, TUG-891 was recently
identified as a GPR120 agonist, which has better selective
potency on human and mice GPR120 than GW9508 and
NCG21 [27, 63]. In our previous work, using TUG-891
to activate porcine GPR120, we found that this agonist
shares the strong response to GPR120 similar to DHA and
ALA [21]. Furthermore, TUG-891 can stimulate intracel-
lular calcium release in a concentration-dependent manner
in 2-day differentiated 3T3-L1 adipocytes [24]. Both in
the ectopically expressing HEK293T cells and the endog-
enously expressing adipocytes, ERK1/2 signal can be well
phosphorylated by TUG-891 treatment [21, 24]. However,
the selectivity of TUG-891 is limited for mice and is still
unknown for rat GPR120. In addition, the orally available
and in vivo tested GPR120 agonist cpdA has been devel-
oped [44]. cpdA mimics the anti-inflammatory effects with
DHA, as Oh etal. reported, and showed a GPR120-medi-
ated anti-inflammation role invivo [44].
It is certain that GPR120 improves glucose uptake and
provides anti-inflammatory/insulin-sensitizing effects.
Uncovering the detailed mechanism of GPR120-n-3 PUFA
will lead to a more direct and potent way to guide the
proper supplementation of fish oil. Especially, in regard
to the anti-inflammatory effect, n-3 PUFA could activate
GPR120 in the hypothalamus in a diet-induced obesity
mice model, and further studies are still needed to shed
light on the role of GPR120 to alleviate inflammation in the
central nervous system (CNS) [78, 79]. As a “druggable”
GPCR, the development of a highly efficient and functional
GPR120 agonist both invitro and in vivo could lead to a
new therapeutic approach for the treatment of systemic
metabolic diseases (Fig.2) [80].
Conclusion andperspective
Over the last decade, recent studies have illustrated that
GPR120 acts as a positive regulator of adipogenesis and
improves glucose uptake in adipocytes, as well as having an
anti-inflammatory effect in ATMs. Its different roles during
the development and metabolism of adipocytes may sug-
gest that the expression and activity of GPR120 is capable
of promoting normal lipid and glucose metabolism (Fig.2).
Therefore, useful approaches to modulating GPR120 activ-
ity may be developed as promising anti-obesity and anti-
diabetic agents. Currently, highly efficient and potent drugs
targeting GPR120 are being developed, though one kind of
agonist, cpdA, shows functional effects both in vivo and
invitro in the regulation of glucose uptake, anti-inflamma-
tory activity, and insulin resistance. Although some impor-
tant issues have been solved to uncover the role of GPR120,
other questions remain to be addressed in the future.
First, some studies demonstrate that the signals trans-
duced from membrane-associated GPR120 into the cells
result in the regulation of the nuclear receptor PPARγ.
What are the specific events in this putative GPR120-
PPARγ pathway? Calcium and ERK1/2 may be two second
messengers that help transduce the signal into the cell, but
GPR120: acritical role inadipogenesis, inflammation, andenergy metabolism inadipose tissue
1 3
the subsequent mechanism that activates PPARγ needs to
be dissected. GPR120 and PPARγ exhibit a similar pocket
structure, and n-3 PUFA can activate both of them, which
means that co-activation might be a model to investigate
in further research, such as in studies of anti-inflammatory
properties. Furthermore, the precise downstream signalling
mediated by GPR120 in brown adipocytes adipogenesis is
worth being uncovered.
Second, as an important endocrine organ, adipose tis-
sue is distributed or intertwined in different organs, such as
muscle, heart, kidney, liver, or even cancer cells. The inter-
actions between adipose tissue and other organs are being
explored and show promise [3]. GPR120 maintains the nor-
mal adipose-liver-muscle interactions, and the dysfunction
of GPR120 may lead to insufficient production of palmi-
toleate (C16:1n7), leading to disorders in lipid metabolism
in the liver and muscle [10]. The systemic role of GPR120
in adipose tissue interactions with other organs needs to be
further verified.
Finally, GPR120 signals via both β-arrestin 2 and
G proteins in the reported studies. As is well known,
diverse proteins are coupled in different biological events.
β-Arrestin2 mediates the anti-inflammatory effect through
internalization with the receptor, while in lipid and glucose
metabolism, two downstream signals, intracellular calcium
mobilization, and phosphorylation of ERK1/2 are the con-
sequence of G protein coupling. However, direct evidence
for the receptor and G protein interaction is lacking. Fur-
thermore, there are few reports about the roles of other pos-
sible protein interactions in these events, such as G protein-
coupled receptor kinase 6 (GRK6) [81, 82]. Thus, it is of
great interest to clarify the precise downstream signals in
different processes. This may help us better understand the
actual function of GPR120 and better design agents for the
treatment of metabolic diseases.
Acknowledgements We apologize to those authors whose excel-
lent work we could not reference directly in this review due to lim-
ited text space. This study was jointly supported by the National
Natural Science Foundation of China (Nos. 31472075 and 31402085);
Hubei Provincial Creative Team Project of Agricultural Science
and Technology (No. 2007-620); the Key Technology Research and
Development Program of Hubei Province (Nos. 2014ABB014 and
2014ABC012).
Compliance with ethical standards
Conflict of interest The authors declare no conflicts of interest.
References
1. Hajer GR, van Haeften TW, Visseren FLJ (2008) Adipose tissue
dysfunction in obesity, diabetes, and vascular diseases. Eur Heart
J 29(24):2959–2971
2. Cannon B, Nedergaard J (2004) Brown adipose tissue: Func-
tion and physiological significance. Physiol Rev 84(1):277–359
3. Rosen ED, Spiegelman BM (2014) What we talk about when
we talk about fat. Cell 156(1–2):20–44
4. Kershaw EE, Flier JS (2004) Adipose tissue as an endocrine
organ. J Clin Endocrinol Metab 89(6):2548–2556
5. Harwood HJ (2012) The adipocyte as an endocrine organ in
the regulation of metabolic homeostasis. Neuropharmacology
63(1):57–75
6. Duplus E, Forest C (2002) Is there a single mechanism for
fatty acid regulation of gene transcription? Biochem Pharma-
col 64(5–6):893–901
7. Lee JH etal (2009) Omega-3 fatty acids: cardiovascular bene-
fits, sources and sustainability. Nat Rev Cardiol 6(12):753–758
8. Scorletti E, Byrne CD (2013) Omega-3 fatty acids, hepatic
lipid metabolism, and nonalcoholic fatty liver disease. Annual
Rev Nutr 33: 231–248.
9. Hirasawa A, Tsujimoto G (2005) Ligand identification and
functional analysis for orphan GPCR GPR120. Yakugaku
Zasshi J Pharm Soc Jpn 125: 122–123
10. Ichimura A etal (2012) Dysfunction of lipid sensor GPR120
leads to obesity in both mouse and human. Nature 483(7389):
350–354
11. Oh DY etal (2010) GPR120 Is an omega-3 fatty acid recep-
tor mediating potent anti-inflammatory and insulin-sensitizing
effects. Cell 142(5):687–698
12. Gotoh C etal (2007) The regulation of adipogenesis through
GPR120. Biochem Biophys Res Commun 354(2):591–597
13. Im DS (2016) Functions of omega-3 fatty acids and FFA4
(GPR120) in macrophages. Eur J Pharmacol 785:36–43
14. Moniri NH (2016) Free-fatty acid receptor-4 (GPR120): Cellu-
lar and molecular function and its role in metabolic disorders.
Biochem Pharmacol 110:1–15
15. Ulven T, Christiansen E (2015) Dietary fatty acids and their
potential for controlling metabolic diseases through activation
of FFA4/GPR120. Annual Rev Nutr 35: 239–263
16. Zhang D, Leung PS (2014) Potential roles of GPR120 and
its agonists in the management of diabetes. Drug Design Dev
Therapy 8: 1013–1027
17. Hirasawa A etal (2005) Free fatty acids regulate gut incretin
glucagon-like peptide-1 secretion through GPR120. Nat Med
11(1):90–94
18. Tanaka T et al (2008) Cloning and characterization of the
rat free fatty acid receptor GPR120: in vivo effect of the
natural ligand on GLP-1 secretion and proliferation of pan-
creatic beta cells. Naunyn Schmiedebergs Arch Pharmacol
377(4–6):515–522
19. Moore K et al (2009) Cloning, expression, and pharmaco-
logical characterization of the GPR120 free fatty acid receptor
from cynomolgus monkey: comparison with human GPR120
splice variants. Comp Biochem Physiol B-Biochem Mol Biol
154(4):419–426
20. Watson SJ, A.J.H. Brown, Holliday ND (2012) Differential sign-
aling by splice variants of the human free fatty acid receptor
GPR120. Mol Pharmacol 81(5):631–642
21. Song TX et al (2015) Cloning and characterization of spliced
variants of the porcine G protein coupled receptor 120. Biomed
Res Int 2015:1–10
22. Tanaka T et al (2008) Free fatty acids induce cholecystokinin
secretion through GPR120. Naunyn Schmiedebergs Arch Phar-
macol 377(4–6):523–527
23. Otto TC, Lane MD (2005) Adipose development: From stem cell
to adipocyte. Crit Rev Biochem Mol Biol 40(4):229–242
24. Song TX, et al. (2016) GPR120 promotes adipogenesis through
intracellular calcium and extracellular signal-regulated kinase
1/2 signal pathway. Mol Cell Endocrinol 434(C): 1–13.
T.Song et al.
1 3
25. Ichimura A, Hara T, Hirasawa A (2014) Regulation of energy
homeostasis via GPR120. Front Endocrinol 5:111
26. Quesada-Lopez T et al (2016) The lipid sensor GPR120 pro-
motes brown fat activation and FGF21 release from adipocytes.
Nat Commun 7:13479
27. Hudson BD et al (2013) The pharmacology of TUG-891, a
potent and selective agonist of the free fatty acid receptor 4
(FFA4/GPR120), demonstrates both potential opportunity and
possible challenges to therapeutic agonism. Mol Pharmacol
84(5):710–725
28. Gao B etal (2015) GPR120: A bi-potential mediator to modulate
the osteogenic and adipogenic differentiation of BMMSCs. Sci
Rep 5:14080
29. Kim J etal (2016) Eicosapentaenoic acid potentiates brown ther-
mogenesis through FFAR4-dependent up-regulation of miR-30b
and miR-378. J Biol Chem 291(39):20551–20562
30. Gim HJ et al (2013) Design and synthesis of alkoxyindolyl-
3-acetic acid analogs as peroxisome proliferator-activated
receptor-gamma/delta agonists. Bioorganic Med Chem Lett
23(2):513–517
31. Suzuki T etal (2008) Identification of G protein-coupled recep-
tor 120-selective agonists derived from PPAR gamma agonists. J
Med Chem 51(23):7640–7644
32. Hudson BD etal (2014) The molecular basis of ligand interac-
tion at free fatty acid receptor 4 (FFA4/GPR120). J Biol Chem
289(29):20345–20358
33. Bost F etal (2005) The role of MAPKs in adipocyte differentia-
tion and obesity. Biochimie 87(1):51–56
34. Prusty D etal (2002) Activation of MEK/ERK signaling pro-
motes adipogenesis by enhancing peroxisome proliferator-acti-
vated receptor gamma (PPAR gamma) and C/EBP alpha gene
expression during the differentiation of 3T3-L1 preadipocytes. J
Biol Chem 277(48):46226–46232
35. Neal JW, Clipstone NA (2002) Calcineurin mediates the cal-
cium-dependent inhibition of adipocyte differentiation in 3T3-L1
cells. J Biol Chem 277(51):49776–49781
36. Shi H etal (2000) Role of intracellular calcium in human adipo-
cyte differentiation. Physiol Genomics 3(2):75–82
37. Strissel KJ et al (2007) Adipocyte death, adipose tissue remod-
eling, and obesity complications. Diabetes 56(12):2910–2918
38. Ouchi N etal (2011) Adipokines in inflammation and metabolic
disease. Nat Rev Immunol 11(2):85–97
39. Lumeng CN, Saltiel AR (2011) Inflammatory links between obe-
sity and metabolic disease. J Clin Invest 121(6):2111–2117
40. Weisberg SP etal (2003) Obesity is associated with macrophage
accumulation in adipose tissue. J Clin Invest 112(12):1796–1808
41. Weisberg S etal (2003) Obesity leads to macrophage accumula-
tion in adipose tissue. Obes Res 11:A6–A7
42. Xu HY et al (2003) Chronic inflammation in fat plays a crucial
role in the development of obesity-related insulin resistance. J
Clin Invest 112(12):1821–1830
43. Harkins JM etal (2004) Expression of interleukin-6 is greater in
preadipocytes than in adipocytes of 3T3-L1 cells and C57BL/6 J
and ob/ob mice. J Nutr 134(10):2673–2677
44. Oh DY etal (2014) A Gpr120-selective agonist improves insu-
lin resistance and chronic inflammation in obese mice. Nat Med
20(8):942–947
45. Liu YQ et al (2014) The fish oil ingredient, docosahexaenoic
acid, activates cytosolic phospholipase A(2) via GPR120 recep-
tor to produce prostaglandin E-2 and plays an anti-inflammatory
role in macrophages. Immunology 143(1):81–95
46. Li XZ, Yu Y, Funk CD (2013) Cyclooxygenase-2 induc-
tion in macrophages is modulated by docosahexaenoic acid
via interactions with free fatty acid receptor 4 (FFA4). Faseb J
27(12):4987–4997
47. Buysschaert M et al (2000) Hyperhomocysteinemia in type 2
diabetes: relationship to macroangiopathy, nephropathy, and
insulin resistance. Diabetes Care 23(12):1816–1822
48. Li Y etal (2008) Homocysteine upregulates resistin production
from adipocytes invivo and invitro. Diabetes 57(4):817–827
49. Li Y etal (2013) Hyperhomocysteinemia promotes insulin resist-
ance by inducing endoplasmic reticulum stress in adipose tissue.
J Biol Chem 288(14):9583–9592
50. Ozcan U etal (2006) Chemical chaperones reduce ER stress and
restore glucose homeostasis in a mouse model of type 2 diabetes.
Science 313(5790):1137–1140
51. Karpe F, Dickmann JR, Frayn KN (2011) Fatty acids, obe-
sity, and insulin resistance: time for a reevaluation. Diabetes
60(10):2441–2449
52. Vieira WA, Sadie-Van Gijsen H, Ferris WF (2016) Free fatty
acid G-protein coupled receptor signaling in M1 skewed
white adipose tissue macrophages. CMLS Cell Mol Life Sci
73(19):3665–3676
53. Iyer A etal (2010) Inflammatory lipid mediators in adipocyte
function and obesity. Nat Rev Endocrinol 6(2):71–82
54. Boden G, Shulman GI (2002) Free fatty acids in obesity and
type 2 diabetes: defining their role in the development of insulin
resistance and beta-cell dysfunction. Eur J Clin Invest 32:14–23
55. Rodriguez-Pacheco F et al (2016) The pro-/anti-inflammatory
effects of different fatty acids on visceral adipocytes are par-
tially mediated by GPR120. Eur J Nutr 1–10. doi:10.1007/
s00394-016-1222-0
56. Rodriguez-Pacheco F et al (2014) Effects of obesity/fatty
acids on the expression of GPR120. Mol Nutr Food Res
58(9):1852–1860
57. Cornall LM etal (2011) Diet-induced Obesity up-regulates the
abundance of GPR43 and GPR120 in a tissue specific manner.
Cell Physiol Biochem 28(5):949–958
58. Trayhurn P (2013) Hypoxia and adipose tissue function and dys-
function in obesity. Physiol Rev 93(1):1–21
59. Krishnan J et al (2012) Dietary obesity-associated Hif1 alpha
activation in adipocytes restricts fatty acid oxidation and energy
expenditure via suppression of the Sirt2-NAD(+) system. Genes
Dev 26(3):259–270
60. Sun K et al (2012) Dichotomous effects of VEGF-A on adipose
tissue dysfunction. Proc Natl Acad Sci USA 109(15):5874–5879
61. Hasan AU et al. (2015) Eicosapentaenoic acid upregulates
VEGF-A through both GPR120 and PPAR gamma medi-
ated pathways in 3T3-L1 adipocytes. Mol Cell Endocrinol
406(C):10–18.
62. Yore MM etal (2014) Discovery of a class of endogenous mam-
malian lipids with anti-diabetic and anti-inflammatory effects.
Cell 159(2):318–332
63. Shimpukade B etal (2012) Discovery of a potent and selective
GPR120 agonist. J Med Chem 55(9):4511–4515
64. Liu D etal (2012) G-protein coupled receptor 120 Is involved in
glucose metabolism in fat cells. Cell Mol Biol 58:1757–1762
65. Chen K etal (2016) Transcription factor C/EBP beta promotes
the transcription of the porcine GPR120 gene. J Mol Endocrinol
56(2):91–100
66. Kajimura S (2017) Adipose tissue in 2016: advances in the
understanding of adipose tissue biology. Nat Rev Endocrinol
13(2):69–70
67. Danforth E Jr (2000) Failure of adipocyte differentiation causes
type II diabetes mellitus? Nat Genet 26(1):13
68. Cao HM et al (2008) Identification of a lipokine, a lipid hor-
mone linking adipose tissue to systemic metabolism. Cell
134(6):933–944
69. Denis GV, Obin MS (2013) ‘Metabolically healthy obesity’: ori-
gins and implications. Mol Aspects Med 34(1):59–70
GPR120: acritical role inadipogenesis, inflammation, andenergy metabolism inadipose tissue
1 3
70. Kloting N etal (2010) Insulin-sensitive obesity. Am J Physiol
Endocrinol Metab 299(3):E506–E515
71. Tang W etal (2011) Thiazolidinediones Regulate Adipose Line-
age Dynamics. Cell Metab 14(1):116–122
72. Gustafson B et al (2013) Restricted adipogenesis in hyper-
trophic obesity the role of WISP2, WNT, and BMP4. Diabetes
62(9):2997–3004
73. Rosell M etal (2014) Brown and white adipose tissues: intrinsic
differences in gene expression and response to cold exposure in
mice. Am J Physiol Endocrinol Metab 306(8):E945–E964
74. Briscoe CP et al (2006) Pharmacological regulation of insulin
secretion in MIN6 cells through the fatty acid receptor GPR40:
identification of agonist and antagonist small molecules. Br J
Pharmacol 148(5):619–628
75. Galindo MM etal (2012) G protein-coupled receptors in human
fat taste perception. Chem Senses 37(2):123–139
76. Mobraten K etal (2013) Omega-3 and omega-6 PUFAs induce
the same GPR120-mediated signalling events, but with differ-
ent kinetics and intensity in Caco-2 cells. Lipids Health Disease
12(1):101
77. Wu Q et al (2013) Identification of G-protein-coupled recep-
tor 120 as a tumor-promoting receptor that induces angiogen-
esis and migration in human colorectal carcinoma. Oncogene
32(49):5541–5550
78. Cintra DE etal (2012) Unsaturated fatty acids revert diet-induced
hypothalamic inflammation in obesity. Plos One 7(1):e30571
79. Wellhauser L, Belsham DD (2014) Activation of the omega-3
fatty acid receptor GPR120 mediates anti-inflammatory actions
in immortalized hypothalamic neurons. J Neuroinflammation
11(1):60
80. Oh DY, Walenta E (2014) Omega-3 fatty acids and FFAR4.
Front Endocrinol 5(4):115
81. Shenoy SK, Lefkowitz RJ (2011) beta-arrestin-mediated recep-
tor trafficking and signal transduction. Trends Pharmacol Sci
32(9):521–533
82. Strachan RT etal (2014) Divergent transducer-specific molecular
efficacies generate biased agonism at a G Protein-coupled recep-
tor (GPCR). J Biol Chem 289(20):14211–14224
... G protein-coupled receptor 120 (GPR120) is distributed in the body's fat and liver. GPR120 affects the expression of glucagon-like peptides and also has a role in regulating adipogenesis, anti-inflammation, and insulin sensitisation [143][144][145]. GPR120 activation inhibits LPS-induced inflammation. ...
Article
Full-text available
Endotoxin is a general term for toxic substances in Gram-negative bacteria, whose damaging effects are mainly derived from the lipopolysaccharides (LPS) in the cell walls of Gram-negative bacteria, and is a strong pyrogen. Obesity is a chronic, low-grade inflammatory condition, and LPS are thought to trigger and exacerbate it. The gut flora is the largest source of LPS in the body, and it is increasingly believed that altered intestinal microorganisms can play an essential role in the pathology of different diseases. Today, the complex axis linking gut flora to inflammatory states and adiposity has not been well elucidated. This review summarises the evidence for an interconnection between LPS, obesity, and gut flora, further expanding our understanding of LPS as a mediator of low-grade inflammatory disease and contributing to lessening the effects of obesity and related metabolic disorders. As well as providing targets associated with LPS, obesity, and gut flora, it is hoped that interventions that combine targets with gut flora address the individual differences in gut flora treatment.
... It shows that obesity is linked to a lipid metabolism issue; there is an indication that 43.2% of people with obesity have hyperlipidemia [93]. An important function of adipose tissue is to regulate energy metabolism [94]. It has been proposed that one of the key strategies for treating obesity is to activate brown adipose tissue (BAT) and produce browning in white adipose tissue (WAT) [95]. ...
Article
Full-text available
Rare ginsenoside compound K (CK) is an intestinal microbial metabolite with a low natural abundance that is primarily produced by physicochemical processing, side chain modification, or metabolic transformation in the gut. Moreover, CK exhibits potent biological activity compared to primary ginsenosides, which has raised concerns in the field of ginseng research and development, as well as ginsenoside-related dietary supplements and natural products. Ginsenosides Rb1, Rb2, and Rc are generally used as a substrate to generate CK via several bioconversion processes. Current research shows that CK has a wide range of pharmacological actions, including boosting osteogenesis, lipid and glucose metabolism, lipid oxidation, insulin resistance, and anti-inflammatory and anti-apoptosis properties. Further research on the bioavailability and toxicology of CK can advance its medicinal application. The purpose of this review is to lay the groundwork for future clinical studies and the development of CK as a therapy for metabolic disorders. Furthermore, the toxicology and pharmacology of CK are investigated as well in this review. The findings indicate that CK primarily modulates signaling pathways associated with AMPK, SIRT1, PPARs, WNTs, and NF-kB. It also demonstrates a positive therapeutic effect of CK on non-alcoholic fatty liver disease (NAFLD), obesity, hyperlipidemia, diabetes, and its complications, as well as osteoporosis. Additionally, the analogues of CK showed more bioavailability, less toxicity, and more efficacy against disease states. Enhancing bioavailability and regulating hazardous variables are crucial for its use in clinical trials.
... It shows that obesity is linked to a lipid metabolism issue; there is an indication that 43.2% of obese people have hyperlipidemia [93]. An important function of adipose tissue is to regulate energy metabolism [94]. It's been proposed that one of the key strategies for treating obesity is to activate brown adipose tissue (BAT) and produce browning in white adipose tissue (WAT) [95]. ...
Preprint
Full-text available
Rare ginsenoside compound K (CK), is an intestinal microbial metabolite with a low natural abundance that is primarily produced by physicochemical processing, side chain modification, or metabolic transformation in the gut. Moreover, CK exhibits potent biological activity compared to primary ginsenosides, which has raised concerns in the field of ginseng research and development as well as ginsenosides-related dietary supplements and natural products. Ginsenosides Rb1, Rb2, and Rc are generally used as a substrate to generate CK via several bio-conversion processes. Current research shows that CK has a wide range of pharmacological actions including boosting osteogenesis, lipid and glucose metabolism, lipid oxidation, insulin resistance, anti-inflammatory, and anti-apoptosis properties. Further research on the bioavailability and toxicology of CK can advance its medicinal application. The purpose of this review is to lay the groundwork for future clinical studies and the development of CK as a therapy for metabolic disorders. Furthermore, the toxicology and pharmacology of CK are investigated as well in this review. The findings indicate that CK primarily modulates signal-ing pathways associated with AMPK, SIRT1, PPARs, WNTs, and NF-kB. It also demonstrates a positive therapeutic effect of CK on nonalcoholic fatty liver disease, obesity, hyperlipidemia, diabetes, and its complications, as well as osteoporosis. Additionally, the analogues of CK showed more bioavailability, less toxicity, and more efficacy against disease states. Enhancing bioavailability and regulating hazardous variables are crucial for its use in clinical trials.
... The relationship between comorbidity and its impact on insulin resistance, a known contributor to obesity-associated metabolic illnesses, is not yet fully understood. However, there have been reports suggesting that comorbidity may ameliorate insulin resistance by interacting with proteins such as the peroxisome proliferator-activated receptor PPARγ and GPR120 (Song et al. 2017). The GPR120 receptor is responsible for recognizing and binding to long-chain fatty acids. ...
Article
Full-text available
Obesity is a cause of comorbid diseases such as type 2 diabetes mellitus, dyslipidemia, hypertension which is based on low-level chronic inflammation. The GPR-120 receptor plays a role in insulin sensitization which is related to diabetes mellitus which is a comorbid obesity. Omega-3 fatty acids are believed to possess anti-inflammatory properties, hence potentially serving as a preventive measure against obesity-related comorbidities. The aim of this study is to do a stability analysis of the binding affinity between nine specific chemicals derived from omega-3 and the active site of the human GPR120 receptor using molecular dynamics simulations. Docking analysis was performed using Discovery Studio Visualizer, AutoDock Tools 1.5.6, and molecular dynamic simulation with AMBER 16. In this study, we used neurotensin 8–13 as a natural ligand to bind with the neurotensin receptor. Based on the neurotensin receptor docking results, the ΔG values for the following compounds are close to the values for neurotensin 8–13 -6.41 kcal/mol; docosahexaenoic acid -8.96 kcal/mol; eicosapentaenoic acid -7.41 kcal/mol; and heneicosapentaenoic acid -6.34 kcal/mol. Neurotensin 8–13 forms hydrogen bonds with TYR146, ARG213, and PHE344 of the neurotensin receptor, whereas docosahexaenoic acid forms hydrogen bonds with TYR146. Meanwhile, the average RMSD fluctuations for each system, namely docosahexaenoic acid, eicosapentaenoic acid, and heneicosapentaenoic acid, were 0.672, 0.437, and 0.650, respectively. The SASA of the neurotensin receptor-ligand complex showed similar fluctuations, with the average values for docosahexaenoic acid, eicosapentaenoic acid, and heneicosapentaenoic acid being 230.40, 229.89, and 230.20 nm ² .
... Nevertheless, the protective effects of MUFA or PUFA intake on diabetes risk may be related to an incretin peptide hormone, glucagon-like peptide-1 (GLP-1). GPR120 is a receptor expressed in the adipose tissue, pro-inflammatory macrophages, and gastrointestinal tract, especially in the enteroendocrine L cells (42). Notably, PUFA can bind with GPR120 to promote the release of GLP-1, which further affects insulin secretion. ...
Article
Full-text available
Objective Unsaturated fatty acids (UFA) may be related to glycometabolism. While associations between UFA intake (especially their subtype) and prediabetes or type 2 diabetes mellitus (T2DM) need to be further studied. In this study, we aimed to evaluate the potential relation of UFA with prediabetes and T2DM. Methods A total of 16,290 adults aged older than 18 years from the National Health and Nutrition Examination Survey (NHANES) from 2005 to March 2020 were included in the present analysis. Dietary intake was assessed by two day, 24-hour dietary recalls and daily intake of total monounsaturated fatty acids (MUFA) and polyunsaturated fatty acids (PUFA); four specific fatty acids of MUFA and seven specific fatty acids of PUFA were calculated. Prediabetes and T2DM were diagnosed by fasting glucose, glycohemoglobin, and self-reported medication or insulin. Rao–Scott modified chi-square tests, the Taylor series linearization method, and multivariable logistic regression analyses were applied to analyze the associations of dietary MUFA and PUFA intake with diabetes risk. Results Of the participants, 44.34% had prediabetes and 13.16% had T2DM patients. From multivariate analysis, we found that intake of MUFA, PUFA, and some subtypes was negatively associated with the risk of prediabetes and T2DM in Americans. Compared with adults in the lowest tertile, those in the highest MUFA (PUFA) tertile had an approximately 50% (49%) and 69% (68%) lower risk of prediabetes and T2DM, respectively. Moreover, the effects of the subtypes of MUFA and PUFA on prediabetes and T2DM were different. Higher intakes of MFA 18:1, MFA 20:1, PFA 18:2, and PFA 18:3 and higher tertile intakes of MFA 16:1 and PFA 20:4 were related to a lower risk of prediabetes and T2DM. Similarly, the effects of MUFA, PUFA, and subtype on prediabetes and T2DM varied among different age groups, being weakened along with age. Conclusion Our study suggested that total MUFA and PUFA intake might be essential in preventing prediabetes and T2DM, especially in Americans. However, this protective effect may decrease with age. Moreover, the effects of the specific UFA on prediabetes and T2DM need further consideration.
Article
Obesity, a burgeoning worldwide health system challenge, is associated with multiple chronic diseases, including diabetes and chronic inflammation. Fatty acid esters of hydroxy fatty acids (FAHFAs) are newly identified lipids with mitigating and anti‐inflammatory effects in diabetes. Increasing work has shown that FAHFAs exert antioxidant activity and enhance autophagy in neuronal cells and cardiomyocytes. We systematically summarized the biological activities of FAHFAs, including their regulatory effects on diabetes and inflammation, antioxidant activity, and autophagy augmentation. Notably, the structure–activity relationships and potential biosynthesis of FAHFAs are thoroughly discussed. FAHFAs also showed potential roles as diagnostic biomarkers. FAHFAs are a class of resources with promising applications in the biomedical field that require in‐depth research and hotspot development, as their structure has not been fully resolved and their biological activity has not been fully revealed.
Article
G protein-coupled receptors (GPCRs) are the largest family of cell surface receptors, with many GPCRs having crucial roles in endocrinology and metabolism. Cryogenic electron microscopy (cryo-EM) has revolutionized the field of structural biology, particularly regarding GPCRs, over the past decade. Since the first pair of GPCR structures resolved by cryo-EM were published in 2017, the number of GPCR structures resolved by cryo-EM has surpassed the number resolved by X-ray crystallography by 30%, reaching >650, and the number has doubled every ~0.63 years for the past 6 years. At this pace, it is predicted that the structure of 90% of all human GPCRs will be completed within the next 5-7 years. This Review highlights the general structural features and principles that guide GPCR ligand recognition, receptor activation, G protein coupling, arrestin recruitment and regulation by GPCR kinases. The Review also highlights the diversity of GPCR allosteric binding sites and how allosteric ligands could dictate biased signalling that is selective for a G protein pathway or an arrestin pathway. Finally, the authors use the examples of glycoprotein hormone receptors and glucagon-like peptide 1 receptor to illustrate the effect of cryo-EM on understanding GPCR biology in endocrinology and metabolism, as well as on GPCR-related endocrine diseases and drug discovery.
Article
Full-text available
Adipose tissues have a central role in energy homeostasis, as they secrete adipokines and regulate energy storage and dissipation. Novel adipokines from white, brown and beige adipocytes have been identified in 2016. Identifying the specific receptors for each adipokine is pivotal for developing greater insights into the fat-derived signalling pathways that regulate energy homeostasis.
Article
Full-text available
The thermogenic activity of brown adipose tissue (BAT) and browning of white adipose tissue are important components of energy expenditure. Here we show that GPR120, a receptor for polyunsaturated fatty acids, promotes brown fat activation. Using RNA-seq to analyse mouse BAT transcriptome, we find that the gene encoding GPR120 is induced by thermogenic activation. We further show that GPR120 activation induces BAT activity and promotes the browning of white fat in mice, whereas GRP120-null mice show impaired cold-induced browning. Omega-3 polyunsaturated fatty acids induce brown and beige adipocyte differentiation and thermogenic activation, and these effects require GPR120. GPR120 activation induces the release of fibroblast growth factor-21 (FGF21) by brown and beige adipocytes, and increases blood FGF21 levels. The effects of GPR120 activation on BAT activation and browning are impaired in FGF21-null mice and cells. Thus, the lipid sensor GPR120 activates brown fat via a mechanism that involves induction of FGF21.
Article
Full-text available
Emerging evidence suggests that n-3 poly unsaturated fatty acids (PUFA) promote BAT thermogenesis. However, underlying mechanisms remain elusive. Here, we hypothesize that n-3 PUFA promote brown adipogenesis by modulating miRNAs. To test this hypothesis, murine brown preadipocytes were induced to differentiate with fatty acids of palmitic (PA), oleate (OA), or eicosapentaenoic acid (EPA). The increase of brown-specific signature genes and oxygen consumption rate (OCR) by EPA were concurrent with upregulation of miR-30b and 378, but not by OA or PA. Next, we hypothesize that free fatty acid receptor 4 (FFAR4), a functional receptor for n-3 PUFA, modulates miR-30b and 378. Treatment of FFAR4 agonist (GW9805) recapitulated the thermogenic activation of EPA by increasing OCR, brown-specific marker genes, and miR-30b and 378, which were abrogated in FFAR4-silenced cells. Intriguingly, addition of miR-30b mimic was unable to restore EPA-induced Ucp1 expression in FFAR4-depleted cells, implicating that FFAR4 signaling activity is required for upregulating brown-adipogenic program. Moreover, blockage of miR-30b or 378 by LNA inhibitors significantly attenuated FFAR4 as well as brown-specific signature gene expression, suggesting the signaling interplay between FFAR4 and miR-30b/378. The association between miR-30b/378 and brown thermogenesis was also confirmed in fish oil-fed C57/BL6 mice. Interestingly, the FFAR4 agonism-mediated signaling axis of FFAR4-miR-30b/378-UCP1 was linked with an elevation of cAMP in brown adipocytes, similar to cold-exposed or fish oil-fed brown fat. Taken together, our work identifies a novel function of FFAR4 in modulating brown adipogenesis partly through a mechanism involving cAMP activation and upregulation of miR-30b and miR-378.
Article
Full-text available
Purpose This study examines whether G-protein coupled receptor 120 (GPR120) is involved in the pro-/anti-inflammatory effects of different types of fatty acids (FAs) in human visceral adipocytes, and whether these effects may be altered in obesity, a state with a chronic inflammation. Methods Pro-/anti-inflammatory effects of palmitic, oleic, linoleic and docosahexaenoic acids on human visceral adipocytes were tested in mature adipocytes from non-obese and morbidly obese (MO) subjects. Also, the effects of these FAs were tested when the GPR120 gene was silenced. Results In adipocytes from non-obese subjects, palmitic and linoleic acids increased TNF-α and IL-6 mRNA expression (p < 0.05), and decreased IL-10 and adiponectin expression (p < 0.05). However, oleic and docosahexaenoic acids (DHA) produced the opposite effect (p < 0.05). In adipocytes from MO subjects, all FAs used increased TNF-α and IL-6 expression (p < 0.05). Palmitic and linoleic acids decreased IL-10 and adiponectin expression (p < 0.05), but oleic acid and DHA did not have significant effects. Only oleic acid increased adiponectin expression (p < 0.05). The effects of FAs on TNF-α, IL-6, IL-10 and adiponectin expression in non-obese and MO subjects were significantly annulled when the GPR120 gene was silenced in visceral adipocytes differentiated from human mesenchymal stem cells. Conclusions FAs are capable of directly acting on visceral adipocytes to modulate differently TNF-α, IL-6, IL-10 and adiponectin expression, with a different and greater effect in MO subjects. These effects are largely annulled when GPR120 expression was silenced, which suggests that they could be mediated by GPR120.
Article
Full-text available
Obesity is associated with the establishment and maintenance of a low grade, chronically inflamed state in the white adipose tissue (WAT) of the body. The WAT macrophage population is a major cellular participant in this inflammatory process that significantly contributes to the pathophysiology of the disease, with the adipose depots of obese individuals, relative to lean counterparts, having an elevated number of macrophages that are skewed towards a pro-inflammatory phenotype. Alterations in the WAT lipid micro-environment, and specifically the availability of free fatty acids, are believed to contribute towards the obesity-related quantitative and functional changes observed in these cells. This review specifically addresses the involvement of the five G-protein coupled free fatty acid receptors which bind exogenous FFAs and signal in macrophages. Particular focus is placed on the involvement of these receptors in macrophage migration and cytokine production, two important aspects that modulate inflammation.
Article
Over the last decade, a subfamily of G protein-coupled receptors that are agonized by endogenous and dietary free-fatty acids (FFA) has been discovered. These free-fatty acid receptors include FFA2 and FFA3, which are agonized by short-chained FFA, as well as FFA1 and FFA4, which are agonized by medium-to-long chained FFA. Ligands for FFA1 and FFA4 comprise the family of long chain polyunsaturated omega-3 fatty acids including α-linolenic acid (ALA), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA), suggesting that many of the long-known beneficial effects of these fats may be receptor mediated. In this regard, FFA4 has gathered considerable interest due to its role in ameliorating inflammation, promoting insulin sensitization, and regulating energy metabolism in response to FFA ligands. The goal of this review is to summarize the body of evidence in regard to FFA4 signal transduction, its mechanisms of regulation, and its functional role in a variety of tissues. In addition, recent endeavors toward discovery of small molecules that modulate FFA4 activity are also presented.
Article
Spin-Transfer Torque RAM (STT-RAM), a promising alternative to SRAM for reducing leakage power consumption, has been widely studied to mitigate the impact of its asymmetrically long write latency. Recently, STT-RAM has been proposed for L1 caches by relaxing the data retention time to improve write performance and dynamic energy. However, as the technology scales down from 65nm to 22nm, the performance of the read operation scales poorly due to reduced sense margins and sense amplifier delays. In this article, we leverage a dual-mode STT memory cell to design a configurable L1 cache architecture termed C1C to mitigate read performance barriers with technology scaling. Guided by application access characteristics discovered through novel compiler analyses, the proposed cache adaptively switches between a high performance and a low-power access mode. Our evaluation demonstrates that the proposed cache with compiler guidance outperforms a state-of-the-art STT-RAM cache design by 9p with high dynamic energy efficiency, leading to significant performance/watt improvements over several competing approaches.