Vous êtes sur la page 1sur 10

TIPS-1235; No.

of Pages 10

Review

Adipokines in health and disease


Mathias Fasshauer and Matthias Blüher
Department of Medicine, University of Leipzig, Liebigstrasse 20, D-04103 Leipzig, Germany

Obesity increases the risk for metabolic, cardiovascular, regulate or modulate different biological processes in tar-
chronic inflammatory, and several malignant diseases get organs, including the brain, liver, muscle, vasculature,
and, therefore, may contribute to shortened lifespan. heart and pancreas, immune system, and others (Figure 2)
Adipokines are peptides that signal the functional status [10,11]. Adipokines may exert specific effects on a variety of
of adipose tissue to targets in the brain, liver, pancreas, biological processes, including: immune response [e.g.,
immune system, vasculature, muscle, and other tissues. adipsin, acylating simulation protein (ASP), and serum
Secretion of adipokines, including leptin, adiponectin, amyloid A3 (SAA3)]; inflammation [e.g., interleukin (IL)-
fibroblast growth factor 21 (FGF21), retinol-binding pro- 1b, -6, -8, and -10, tumor necrosis factor a (TNFa), C-
tein 4 (RBP4), dipeptidyl peptidase 4 (DPP-4), bone mor- reactive protein (CrP), monocyte chemotactic protein-1
phogenetic protein (BMP)-4, BMP-7, vaspin, apelin, and (MCP-1), resistin, progranulin, and chemerin]; glucose
progranulin, is altered in adipose tissue dysfunction and metabolism [e.g., leptin, adiponectin, DPP-4, FGF21, resis-
may contribute to a spectrum of obesity-associated dis- tin, vaspin, and angiopoietin-like protein 8 (Angptl8)];
eases. Adipokines are promising candidates both for insulin sensitivity (e.g., leptin, adiponectin, chemerin,
novel pharmacological treatment strategies and as di- RBP4, and omentin); insulin secretion (e.g., apelin and
agnostic tools, provided that we can develop a better nesfatin-1); blood pressure (e.g., apelin and angiotensino-
understanding of the function and molecular targets of gen); myocardial contractility [e.g., fatty acid binding pro-
the more recently discovered adipokines. tein-4 (FABP-4)]; cell adhesion [e.g., plasminogen activator
inhibitor-1 (PAI-1)]; vascular growth and function [e.g.,
Adipokines vascular endothelial growth factor (VEGF)]; adipogenesis
Obesity significantly increases the risk for metabolic dis- and bone morphogenesis (e.g., BMP-4 and -7); growth [e.g.,
eases [type 2 diabetes mellitus (T2DM), fatty liver disease, insulin-like growth factor-1 (IGF-1), transforming growth
and dyslipidemia], cardiovascular disorders (hypertension, factor b (TGFb), and fibronectin]; lipid metabolism (e.g.,
coronary heart disease, and stroke), diseases of the central soluble CD36 and apelin); lipid accumulation in the liver
nervous system (dementia), obstructive sleep apnea, and (e.g., fetuin-A); regulation of appetite and satiety (e.g.,
different types of cancer [1–3]. The main functions of leptin and vaspin); and other biological processes
adipose tissue include the storage of triglycerides under [11]. Therefore, alterations in adipokine secretion may link
conditions of excess calories and their release during per- obesity to its inflammatory, metabolic, and cardiovascular
iods of fasting, thermoregulation, and mechanical organ comorbidities [3,10]. Importantly, the function, molecular
protection [4,5]. Adipose tissue has also been recognized as targets, and potential clinical relevance of many adipo-
an endocrine organ [4–6]. Adipocytes, precursor, endothe- kines are still not known. Functional characterization, in
lial and immune cells, fibroblasts, and others contribute to particular of the more recently identified adipokines, is a
the release of metabolites, lipids, and bioactive peptides; major task in future adipokine research.
so-called ‘adipokines’ (Figure 1). To identify the complete Our understanding of the mechanisms linking obesity
adipose tissue secretome, several proteomic profiling and adipose tissue dysfunction to metabolic and cardiovas-
approaches have been performed recently [7–9]. Although cular disorders is still incomplete. Alterations in adipokine
the full set of human adipokines is still not entirely char- secretion may contribute to obesity-related diseases. Within
acterized, it has become clear that adipose tissue is a the past few years, the number of newly discovered adipo-
source of more than 600 potentially secretory proteins kines has increased significantly and unraveling adipokine
[7]. Adipokines contribute to the regulation of appetite effects has become a hot topic in obesity research. Here, we
and satiety, fat distribution, insulin secretion and sensi- discuss the role of selected adipokines as promising candi-
tivity, energy expenditure, endothelial function, inflamma- dates for future treatment of obesity and obesity-related
tion, blood pressure, and hemostasis [3,10] (Table 1). diseases. We present research data on how adipose tissue
Within adipose tissue, adipokines have been shown to dysfunction changes the adipokine secretion pattern, and
modulate adipogenesis, immune cell migration into adi- focus on the potential of leptin, adiponectin, FGF21, BMP-4,
pose tissue, and adipocyte metabolism and function BMP-7, vaspin, apelin, DPP-4, TNFa, and IL-1b as candi-
[3,11,12] (Figure 2). At the systemic level, adipokines dates for novel pharmacological treatment strategies.

Corresponding author: Blüher, M. (bluma@medizin.uni-leipzig.de).


Keywords: adipokines; obesity; adipose tissue; type 2 diabetes mellitus. Adipose tissue function determines the adipokine
0165-6147/
secretion profile
ß 2015 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.tips.2015.04.014 In overweight and obese individuals, genetic and environ-
mental factors result in a chronic positive energy balance
Trends in Pharmacological Sciences xx (2015) 1–10 1
TIPS-1235; No. of Pages 10

Review Trends in Pharmacological Sciences xxx xxxx, Vol. xxx, No. x

Secretory cells of
adipose ssue
Cytokine and cytokine-
like proteins Adipokines
Lepn
TNFα
Adiponecn
IL-6
Visfan/Nampt/PBEF
MCP-1
Vaspin
Resisn • Adipocytes RBP4
Progranulin • Precursor cells FGF21
• Endothelial cells BMPs
• Macrophages Nesfan-1
Proteins of the
• Foam cells Cathepsins
fibrinolyc system
• Neutrophils Apelin
PAI-1
• Lymphocytes Omenn
Tissue factor
• Fibroblasts Lipocalin
• Others and hundreds more
Complement and
complement-related proteins
Adipsin Proteins of RAS
Complement factor B Angiotensinogen
ASP
CTRPs Endocannabinoids and
Lipid transport other lipids
Enzymes Apolipoprotein E Anandamide
DPP-4 Cholesterol ester transfer protein 2-AG
Lipoprotein lipase Free fay acids
TRENDS in Pharmacological Sciences

Figure 1. Factors released or secreted by adipose tissue. Adipocytes, immune cells, fibroblasts, endothelial cells, and others contribute to the release of metabolites, lipids,
and adipokines. Examples of adipose tissue-derived molecules are provided here. Abbreviations: 2-AG, 2-Arachidonoylglycerol; ASP, acylating simulation protein; BMPs,
bone morphogenetic proteins; CTRPs, C1q/TNF-related proteins; FGF21, fibroblast growth factor 21; MCP-1, monocyte chemotactic protein-1; PAI-1, plasminogen activator
inhibitor-1; RAS, renin angiotensin system; RBP4, retinol binding protein 4. Modified from [3,5,6].

Genec and environmental factors that leads to weight gain. With increasing body weight,
energy imbalance adipose tissue changes its size, distribution, cellular com-
position, and function (Figure 3). The expansion of adipose
Adipose ssue size, cellular composion, and funcon
tissue significantly influences adipocyte biology and may
lead to either the physiologic response of adipose tissue or
to impaired adipose tissue function (Figure 3). In the latter
Impaired
i
(adipocyte hypertrophy, ectopic
case, individuals develop adipocyte hypertrophy (rather
Physiologic
fat accumulaon, and adipose than hyperplasia), ectopic fat deposition, hypoxia, and
ssue inflammaon) chronic stress in adipose tissue, which subsequently causes
an adverse adipokine secretion profile [4] (Figure 3). Hy-
pertrophy of adipocytes is considered a key event associat-
Normal Adverse ed with a loss of insulin sensitivity in both lean and obese
adipokine adipokine
conditions [4,13,14]. Individuals with larger adipocytes
secreon secreon
typically have elevated proinflammatory factors, including
leptin, IL-6, IL-8, and monocyte chemotactic protein 1
Metabolic, inflammatory,
Health (MCP-1) [15], reduced levels of the insulin sensitivity-
and cardiovascular
diseases, cancer related adipokine adiponectin and anti-inflammatory IL-
10 [4,15,16], as well as increased basal and catecholamine-
TRENDS in Pharmacological Sciences
stimulated lipolysis [17]. Adipocyte hypertrophy most like-
Figure 2. Local and systemic effects of adipokines. Within adipose tissue ly initiates alterations in adipose tissue, which predisposes
(representative histologic slide of human omental adipose tissue), adipokines adipose tissue to immune cell infiltration and an adverse
contribute to the regulation of adipogenesis, immune cell migration, adipocyte
metabolism, triglyceride storage, and others. At the whole-body level, adipokines
(proinflammatory, diabetogenic, and atherogenic) adipo-
are important regulators of appetite and satiety, energy expenditure, kine secretion pattern [4]. In addition, adipokine secretion
inflammation, blood pressure, hemostasis, endothelial function influence, insulin may be influenced by fat distribution or may contribute to
sensitivity, and energy metabolism in insulin-sensitive tissues, such as liver,
muscle, and fat, as well as insulin secretion in pancreatic b cells. Modified from
distinct fat distribution subtypes [18]. Several adipokines,
[10,11]. including leptin, IL-6 [19], RBP4 [20], chemerin [21],
2
TIPS-1235; No. of Pages 10

Review Trends in Pharmacological Sciences xxx xxxx, Vol. xxx, No. x

Table 1. The role of selected adipokines in health and diseasea


Adipokine Main actions
Adiponectin Improves insulin sensitivity; antidiabetic, antiatherogenic, and anti-inflammatory
Adipsin Activates the alternative complement pathway
Angiopoietin-like protein 8 Promotes pancreatic b cell proliferation; improves glucose tolerance
Apelin Inhibits insulin secretion
BMP-4 Regulates adipogenic precursor cell commitment and differentiation
BMP-7 Stimulates brown adipogenesis; reduces food intake; increases energy expenditure
Cathepsins S, L, K Regulate glucose metabolism and adipose tissue mass
Chemerin Chemoattractant protein; regulates adipogenesis
Clusterin Promotes tumor progression and angiogenesis
DPP-4 Degrades GIP and GLP-1; inhibitors in clinical use for T2DM
FABP-4 Associated with increased T2DM risk and impaired myocardial contractility
Fetuin-A Reflects liver fat content; associated with lipid-induced inflammation and insulin resistance;
promotes cancer progression
FGF21 Stimulates glucose uptake into adipocytes; increases thermogenesis, energy expenditure, and fat utilization;
improves glucose and lipid metabolism
Gremlin-1 Inhibits BMP-4 and BMP-7
IL-1b Proinflammatory
IL-6 Proinflammatory
Leptin Satiety signal; regulates appetite, food intake, locomotor activity, energy expenditure, fertility, and other
processes
Lipocalin 2 Related to insulin resistance and inflammation
MCP-1 Chemoattractant protein; adipose tissue inflammation
Nesfatin-1 Direct glucose-dependent insulinotropic effect on b cells
Omentin Anti-inflammatory; insulin sensitizing
Progranulin Chemoattractant protein; neurodegenerative diseases; adipose tissue inflammation
RBP4 Related to insulin resistance, visceral fat distribution, and dyslipidemia
Resistin Related to obesity, insulin resistance, and inflammation
TGFb Regulates cell proliferation, differentiation, and apoptosis
Tissue inhibitor of matrix Decreases adipogenesis; impairs glucose tolerance
metalloproteinase-1
TNFa Proinflammatory
Vaspin Serine protease inhibitor; decreases food intake; improves hyperglycemia
VEGF Stimulates angiogenesis in adipose tissue
Visfatin/PBEF/Nampt Nampt-mediated systemic NAD biosynthesis is critical for b cell function
Wnt1 inducible signaling Regulates adipogenesis and adipose tissue inflammation
pathway protein 1
a
Modified from [3,5,10].

progranulin [22], and others, are differentially expressed [body mass index (BMI) >40 kg/m2] could be clearly dis-
in various fat compartments and correlate with obesity- tinguished from age-, gender-, and BMI-matched insulin-
related traits, further supporting their role as potential resistant metabolically healthy obese patients by a normal
mediators of metabolic alterations associated with a dis- adipokine pattern [16]. In the future, adipokines may offer
tinct fat distribution [18,23]. The chronically altered adi- new opportunities for the pharmacotherapy of obesity and
pokine secretion may not only reflect adipose tissue obesity-related diseases and as diagnostic tools [10].
dysfunction, but also contribute to the development of An in-depth discussion of most adipokines is beyond the
metabolic, cardiovascular, inflammatory, and malignant scope of this overview. Therefore, we focus here on selected
diseases (Figure 3). Moreover, using an unbiased hierar- adipokines that are currently used for pharmacotherapy
chical clustering approach, two major adipokine clusters (leptin) or have the potential to be used as therapeutic tools
were recently identified that are related to either body fat (adiponectin, FGF21, BMP-7, vaspin, and apelin) or tar-
mass and inflammation or a combined trait of insulin gets (DPP-4, IL-1b, TNFa, and BMP-4).
sensitivity, glucose, and lipid metabolism [23]. In these
analyses, it was found that even including 20 different Leptin
adipokine serum concentrations in a T2DM prediction Although the endocrine function of adipose tissue has been
model did not reach the sensitivity and specificity of clas- established by observations that adipsin/complement fac-
sical parameters [e.g., hemoglobin (Hb) A1c, homeostatic tor D [24–26] and sex steroids [27] are secreted from
model assessment-insulin resistance (HOMA-IR)] for the adipose tissue, the discovery of leptin in 1994 [28] could
diagnosis of the disease [23], suggesting that measurement be considered the initial milestone for adipokine research
of circulating adipokines alone is currently not clinically [10]. Since then, leptin has represented the prototype for
useful for the diagnosis of metabolic diseases. By contrast, all subsequently identified adipocyte-secreted hormones
individuals who remain insulin sensitive despite obesity [10]. The cloning of leptin also marked the identification
3
TIPS-1235; No. of Pages 10

Review Trends in Pharmacological Sciences xxx xxxx, Vol. xxx, No. x

Appete
Saety
Energy expenditure
Acvity
Immune system
Immune cell aracon Blood pressure
Differenaon Endothelial funcon
Systemic inflammaon Heart muscle contraclity
Wound healing Smooth muscle cells

Insulin sensivity
Lipid accumulaon
Hepatokine secreon
Lipid metabolism
Growth factors

Insulin secreon
Glucagon secreon
Insulin sensivity

Insulin sensivity
Myokine secreon
Lipid storage
Triglyceride storage
Insulin sensivity
Glucose and lipid transport
Adipokine secreon
Differenaon
Cell growth Resorpon
Fat distribuon Incren secreon
Browning

TRENDS in Pharmacological Sciences

Figure 3. Model of the development of either normal or adverse adipokine secretion. As a result of the interaction between genetic and environmental factors, a positive
energy balance affects the size, distribution, cellular composition, and function of adipose tissue. As long as these characteristics of adipose tissue are regulated in a
physiologic manner, adipokine secretion is normal and may contribute to whole-body homeostasis and ‘health’. However, typically a chronic positive energy balance leads
to impaired adipose tissue function, characterized by adipocyte hypertrophy, adipose tissue inflammation, and ectopic fat deposition, which lead to an adverse adipokine
secretion pattern and subsequently contribute to metabolic, cardiovascular, and inflammatory diseases, and cancer.

of the mechanism underlying the extremely obese pheno- leptin treatment of obese patients had only a little effect on
type of the ob/ob mouse model, which carries a mutation in weight loss; both high leptin doses or treatment with
the ob gene subsequently leading to leptin deficiency pegylated leptin once weekly did not cause significant
[28]. For the rare cases of monogenetically inherited leptin weight loss, but did increase the risk for adverse effects
deficiency in humans, recombinant leptin is now available of recombinant leptin therapy [33,34]. The most likely
for compassionate use and leads to significant weight loss reason for the lack of leptin effects on body weight reduc-
[29,30]. Leptin is a 16-kDa protein of 167 amino acids, tion is the development of central leptin resistance or
which circulates both in its free form and bound to proteins. tolerance in obese patients with already very high circu-
Circulating leptin is directly proportional to body fat mass lating leptin levels [10,11]. This hypothesis is further
[31]. Leptin signals via the leptin receptor on target cells, is supported by clinical studies demonstrating that leptin
secreted from adipocytes, and has an important role in the given in combination with amylin (pramlintide), which is
regulation of satiety, appetite, food intake, reproductive co-secreted with insulin from pancreatic b cells and may
function, fertility, puberty, activity, energy expenditure, improve glycemic control as well as leptin sensitivity, is
atherogenesis (reviewed in [10,31]), and fetal growth effective to induce weight loss [35,36]. Recently, a random-
[32]. In the hypothalamus, leptin increases anorexigenic ized clinical trial using a combination of pramlintide with
and decreases orexigenic peptide synthesis, subsequently metreleptin, a synthetic analog of leptin, in obese patients
resulting in reduced appetite [31]. was terminated, because of adverse events, such as anti-
In addition, leptin may exert insulin-sensitizing effects body generation and skin reactions [11,37]. However, for
and is considered an important regulator of b cell mass and the treatment of lipodystrophy, metreleptin is already an
survival (reviewed in [3]). Given its appetite-reducing approved pharmacotherapy in the USA and Japan, and
function in normal or low circulating leptin states, recom- further indications, such as hypothalamic amenorrhea or
binant leptin has been developed as a weight loss phar- Rabson–Mendenhall syndrome, are currently under con-
macotherapy for obesity [10,11,29]. Against expectations, sideration [10,11,38].
4
TIPS-1235; No. of Pages 10

Review Trends in Pharmacological Sciences xxx xxxx, Vol. xxx, No. x

To date, leptin is the best example of a successful path Cancer and Nutrition (EPIC) Potsdam cohort [51]. Low
from its discovery to the clinical application of an adipo- circulating adiponectin has been shown to predict T2DM
kine. even beyond the associations with age, gender, fat distri-
bution, BMI, smoking, exercise, alcohol consumption, edu-
Adiponectin cation, and HbA1c [51]. In accordance with these
Adiponectin was discovered in 1995, shortly after leptin, epidemiologic data, it was recently shown that, together
and has been characterized in mice as the mRNA tran- with immune cell infiltration into visceral adipose tissue,
script most highly expressed in adipocytes [39–42]. Adipo- adiponectin is the strongest predictor of insulin sensitivity
nectin is a 30-kDa protein of 244 amino acids and signals in metabolically healthy obese individuals [16].
via at least two adiponectin receptors, AdipoR1 and Adi- Taken together, adiponectin and adiponectin-pathways
poR2, on target cells [39]. It has become a biochemical are promising candidates for future development in both
paradigm of proteins that are secreted without using the the pharmacotherapy and prediction of cardiometabolic
canonical secretory pathway. Adiponectin is almost exclu- diseases.
sively produced by adipocytes and has gained a lot of
interest due to its insulin sensitizing, anti-inflammatory, FGF21
and antiapoptotic properties [3,32,39]. Moreover, adipo- FGF21 (23 kDa, 209 amino acids) is a member of the FGF
nectin enhances insulin secretion by stimulating both the superfamily that is produced by the liver, adipose tissue,
expression of the insulin gene and exocytosis of insulin and skeletal muscle [52]. Through direct effects on specific
granules. Adiponectin also acts in the brain to increase FGF receptors, FGF21 together with the cofactor b-Klotho
energy expenditure and thereby may promote weight loss has been shown to exert glucose- and lipid-lowering as well
[43]. In clinical studies, circulating adiponectin is indepen- as thermogenic effects [52–55]. In animal studies, recom-
dently and negatively related to facets of the metabolic binant FGF21 reduced hyperglycemia along with addition-
syndrome, including insulin resistance, body weight, blood al beneficial metabolic effects [52]. These FGF21 effects
pressure, and serum lipids [3,32]. could be due, at least in part, to its role in promoting
Post-translational adiponectin modifications result in browning of white adipose tissue and in adaptive thermo-
secreted oligomers of 90-kDa trimers, which are found in genesis [55]. In humans, similar effects were shown in a
the circulation as 180-kDa hexamers [low molecular randomized, placebo-controlled, double-blind, proof-of-con-
weight (LMW)] and 18- to 36-mers [high molecular weight cept trial on the effects of LY2405319 (an FGF21 variant)
(HMW)] [39,43]. HMW adiponectin correlated better with in obese patients with T2DM [53]. Over a treatment period
systemic insulin sensitivity compared with LMW adipo- of 28 days, LY2405319 significantly improved body weight,
nectin in both mouse and human studies [39,43]. fasting insulin, adiponectin, dyslipidemia [including
The molecular actions of adiponectin suggest that the decreases in low-density lipoprotein (LDL) cholesterol
molecule itself or agonists of its receptors have potential for and triglycerides, and increases in high-density lipoprotein
the treatment of obesity and its comorbidities [39]. Indeed, (HDL) cholesterol], and a shift to a potentially less athero-
in rodent models, adiponectin administration improved genic apolipoprotein concentration profile [53]. In contrast
insulin sensitivity and glucose metabolism, increased in- to FGF21 overexpression studies in transgenic mice or of
sulin secretion, and reduced body weight [39,44]. Very recombinant FGF21 treatment in humans, there was only
recently, it was demonstrated that so-called ‘AdipoRon’, a trend for a glucose-lowering effect of LY2405319 in the
an orally active, synthetic small-molecule adiponectin re- clinical study [53].
ceptor agonist, significantly improved insulin sensitivity Counterintuitively to data from FGF21 treatment or
and glucose tolerance in mice [45]. In high fat-fed db/db overexpression studies, FGF21 serum concentrations were
mice, treatment with AdipoRon extended longevity [45] elevated in states of insulin resistance and obesity in
supporting previous suggestions that high adiponectin clinical studies [56]. This may suggest either the develop-
serum concentrations are associated with prolonged life ment of FGF21 resistance in insulin-resistant states or an
span in obesity [46]. Of note, adiponectin levels have been adaptive response to endogenously elevated circulating
shown to inversely correlate with obesity, visceral fat FGF21 concentrations.
distribution, T2DM, and other obesity-related diseases Although treatment with this FGF21 variant shows
[32,39,43]. Therefore, in addition to its potential future promising results for potential future clinical use, safety
clinical importance as a therapeutic tool, adiponectin concerns associated with targeting the FGF21 pathway
represents a promising and easily detectable stable mark- have emerged [57].
er for a variety of diseases [10]. However, in large longi- Mice with a transgenic overexpression of FGF21 have
tudinal studies, higher adiponectin concentrations have reduced body length, most likely due to a decrease in bone
been suggested as either a positive marker for all-cause mass [57,58]. However, FGF21 transgenic mice have in-
mortality, heart failure, and coronary artery disease creased longevity and remain apparently healthy through-
[47,48] or a negative predictor of cardiovascular events out their lifespan [59]. Data from several FGF21
[49], whereas other studies did not find any association transgenic and knockout mouse models led to the hypoth-
between circulating adiponectin and cardiovascular dis- esis that FGF21 is a critical regulator of bone turnover
eases [50]. By contrast, higher adiponectin levels were [57]. In the context of potentially harmful adverse effects
associated with a substantially reduced risk of developing on the skeletal system, it remains an open question wheth-
T2DM in 27 548 apparently healthy participants of the er this might have negative consequences for the potential
population-based European Prospective Investigation into clinical use of FGF21-based treatment strategies.
5
TIPS-1235; No. of Pages 10

Review Trends in Pharmacological Sciences xxx xxxx, Vol. xxx, No. x

Regardless of its future clinical use, the FGF21 drug Vaspin


discovery path from identification in a functional in vitro Visceral adipose tissue-derived serpin (vaspin) was identi-
screen, to the evaluation of its utility in patients [57], may fied as a member of the serine protease inhibitor family.
serve as an example of successful translational adipokine Vaspin (47 kDa, 415 amino acids) was found to be highly
research. expressed in visceral adipose tissue of Otsuka Long-Evans
Tokushima Fatty (OLETF) rats at the age when obesity
BMP-4 and -7 and insulin plasma concentrations peaked [70]. It is not yet
BMPs are members of the TGFb superfamily and regulate known whether vaspin circulates in its free form or bound
several processes of organogenesis, patterning during em- to proteins. Elevated vaspin serum concentrations are
bryonic development, brown and white adipogenesis, and associated with obesity, as well as impaired insulin sensi-
energy expenditure [60–63]. BMP-4 is a 46-kDa protein tivity, fitness level, and leptin serum concentrations
and BMP-7 a 49-kDa protein circulating bound to albumin [71]. In subsequent studies, it became clear that vaspin
and other proteins. Both BMPs are expressed in different expression is not restricted to adipose tissue and can be
tissues (e.g., placenta, thyroid, skin, adipose tissue, and detected in the skin, stomach, and rodent hypothalamus
digestive tract) and signal via two specific BMP receptors, and pancreatic islets [72,73]. Recombinant vaspin admin-
BMPR1 and BMPR2, on target cells. Adipose tissue ex- istration to obese mice improved insulin sensitivity and
pression and/or serum concentrations of different BMPs glucose tolerance [71,73]. In addition, it was shown that
are associated with obesity and fat distribution [18]. More- both peripheral and intracerebroventricular vaspin treat-
over, it has been recently demonstrated that BMP-7 has a ment of different mouse models caused a sustained glucose
role in appetite regulation [63]. Systemic treatment of diet- lowering and reduction of food intake [73]. These beneficial
induced obese mice with BMP-7 led to a significant reduc- vaspin effects could be mediated by its ability to inhibit
tion in body weight and improvement of metabolic param- kallikrein 7 as one target protease [74]. Importantly, at
eters, most likely due to increased energy expenditure and least in vitro, kallikrein 7 is able to cleave and degrade
decreased food intake [63]. The reduction in appetite was insulin in the A and B chain. Vaspin inhibits kallikrein 7 by
also observed in leptin-deficient ob/ob mice, suggesting a classical serpin mechanism with high specificity in vitro
that the action of BMP-7 on food intake is independent of and thereby may contribute to reduced insulin degradation
leptin [63]. Of note, human recombinant BMP-7 is already in the circulation [74]. Moreover, vaspin-kallikrein 7 com-
available under the brand name ‘OP1’, which is used in the plexes have been detected in human plasma and both
context of surgical procedures to aid bone fusion after proteins are co-expressed in murine pancreatic b cells.
fractures [64]. So far, no local or systemic adverse events Therefore, improved glucose metabolism upon vaspin
have been reported from this clinical application treatment could be mediated by increased insulin plasma
[64]. Therefore, it is not unlikely that recombinant BMP- concentrations [74]. Further studies are required to trans-
7 may also enter the development path as a treatment of late the promising data from vaspin treatment studies in
obesity or metabolic diseases. animal models into a potential human application.
BMP-7 may not be the only BMP with a potential future
clinical application. It has been recently shown that dif- Apelin
ferentiated human adipocytes can promote adipogenesis Apelin, a 36 amino-acid peptide (approximately 1.5 kDa)
via endogenous BMP-4 activation, a process that may be and the endogenous ligand of the G protein-coupled recep-
counterbalanced by the expression of the BMP-4 (and -7) tor APLNR, exists in several active forms [32,75,76]. In
inhibitor Gremlin-1 [65]. BMP-4 is secreted by differenti- addition to its role as an adipokine, apelin is expressed in
ated preadipocytes and adipocytes, with higher expression the central nervous system (with particularly high expres-
in larger adipocytes [65]. BMP-4 alone, and/or suppression sion in the hypothalamus), heart, skeletal muscle, and
of Gremlin-1, causes increased peroxisome proliferator- stomach [3,76]. It has been suggested to contribute to
activated receptor g (PPARg) expression and drives pre- the regulation of glucose metabolism, lipolysis, blood pres-
adipocytes towards a beige/brown adipocyte phenotype sure, cardiovascular and fluid homoeostasis, food intake,
[66]. Recently, brown adipose tissue gained a lot of scien- cell proliferation, and angiogenesis [75–80]. In particular,
tific interest with the notion that its occurrence in adult the ability of apelin to decrease lipolysis and induce vaso-
humans is associated with lower BMI, younger age, and dilation with subsequent reduction in blood pressure may
normal glucose metabolism [67–69]. Therefore, there is open new therapeutic avenues [78–80]. Mice with a dis-
growing scientific interest in transforming white into ruption of the apelin gene did not exhibit metabolic altera-
brown adipose tissue as a novel strategy to treat obesity tions, but developed a dysfunction of cardiac contractility
and its related metabolic diseases. BMP-4-related mecha- with aging [81]. Most clinical studies found an association
nisms could represent a future tool for this therapeutic of higher circulating apelin with obesity and insulin resis-
strategy. In hypertrophic obesity, adipose precursor cells tance [79,82]. One potential mechanistic link between
are resistant to BMP-4, which may contribute to the limit- elevated circulating apelin and deterioration of glucose
ed expandability of adipose tissue and, subsequently, to metabolism could be provided by experimental evidence
obesity-related metabolic diseases [16,65]. Taken together, that apelin inhibited insulin secretion in mice [83]. In
these data underscore the importance of BMPs in regulat- insulin-resistant and obese mouse models, administration
ing food intake, energy expenditure, and adipogenesis, and of recombinant apelin had significant beneficial effects on
suggest BMPs as a new therapeutic approach to treat glucose tolerance and glucose utilization [83]. These data
obesity and its comorbidities. suggest that, in obesity and diabetes, increased circulating
6
TIPS-1235; No. of Pages 10

Review Trends in Pharmacological Sciences xxx xxxx, Vol. xxx, No. x

apelin is a symptom of apelin resistance or represents an dysfunction. In future studies, it should be investigated
adaptation of the body to elevated endogenous apelin whether treatment with DPP-4 inhibitors may improve
levels. However, there is a need to investigate the meta- adipose tissue function and whether such treatment leads
bolic effects of apelin administration in both physiological to a better response in those patients with T2DM and
and pathological situations in humans. higher DPP-4 activity and serum concentration.

DPP-4 TNFa
Intake of nutrients may stimulate the release of the incre- TNFa (25 kDa, 233 amino acids) is a proinflammatory adi-
tin hormones, including glucose-dependent insulinotropic pokine that is expressed in monocytes and macrophages
polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) and has a central role in inflammation and autoimmune
[84,85]. Incretins have been shown to enhance glucose- diseases (reviewed in [32]). TNFa can bind two receptors,
dependent insulin secretion and inhibit glucagon secre- TNFR1 and TNFR2. Although adipose tissue TNFa expres-
tion, which together may reduce hepatic gluconeogenesis sion in humans is relatively low, it correlates with obesity
[85]. The effect of incretins is significantly reduced in and decreases with weight loss [92–94]. TNFa has been
patients with T2DM and contributes to impaired insulin shown to directly impair both insulin signaling in insulin-
secretion and chronic hyperglycemia [85]. Under normal sensitive tissues and insulin secretion [92,93]. Therefore,
physiological conditions, DPP-4 rapidly degrades GIP and inhibiting TNFa action may exert beneficial metabolic
GLP-1 [86]. Inhibition of DPP-4 is a therapeutic option to effects. Indeed, neutralization of TNFa by antibodies as well
lower hyperglycemia in patients with T2DM. DPP-4 inhi- as genetic ablation of the TNFa gene had insulin-sensitizing
bitors are a relatively new class of oral glucose-lowering effects in various rodent models obesity and diabetes
drugs, and exert their hypoglycemic effect indirectly by [94–96]. Moreover, in several independent animal models,
increasing plasma concentration, and the duration and treatment with TNFa antibodies was shown to reduce in-
action of incretins [85,87]. Currently, the DPP4-inhibitors flammation, improve fatty liver disease [93], and protect
alogliptin, sitagliptin, linagliptin, saxagliptin, and vilda- against diet-induced obesity and insulin resistance [97].
gliptin are in clinical use as antidiabetes drugs to improve However, these promising data could not be translated
glycemic control by stimulating glucose-induced pancreatic into a clinically relevant treatment option in humans. Of
insulin secretion and suppressing glucagon production note, in patients with spondyloarthritis, anti-TNFa thera-
[85–87]. py contributed to an early significant increase in both
DPP-4 is an 88-kDa protein that is expressed in glan- visceral and subcutaneous abdominal adipose tissue after
dular cells in salivary gland, prostate, seminal vesicles, 1–2 years of treatment [98]. Moreover, in a prospective,
endometrium, renal tubules, and small intestine and de- randomized, double-blind, placebo-controlled pilot study of
cidual cells. Recently, adipose tissue was shown to be an insulin-resistant obese men, chronic TNFa neutralization
additional source of circulating DPP-4 [88]. Compared with by infliximab did not improve insulin resistance and ques-
healthy lean individuals, patients with obesity and insulin tioned the suggested causative link between adiposity and
resistance have higher DPP-4 expression in (visceral) adi- insulin resistance, at least in human disease [99].
pose tissue, which may contribute to increased fat-derived
DPP-4 secretion and higher circulating DPP-4, particular- IL-1b
ly in individuals with high visceral fat mass [18,89]. Given IL-1b (17 kDa, 153 amino acids) is a proinflammatory
that DPP-4 is an important drug target for the treatment of cytokine that is not only produced by immune cells, but
T2DM, it could be hypothesized that enhanced secretion of also secreted from adipose tissue. It circulates mainly
DPP-4 in patients with adverse fat distribution [18] may bound to albumin and signals via two IL-1 receptors (IL-
contribute to the development of impaired insulin sensi- 1R1 and IL-1R2) on target cells. Importantly, IL-1b may
tivity in obesity. However, so far there is no evidence that have a role in inflammatory pancreatic b cell destruction
increased DPP-4 levels from adipose tissue affect endoge- and apoptosis, thus contributing to the development of
nous GLP-1 levels or that DPP-4 inhibitors might be more type 1 diabetes mellitus [100]. It has been suggested that
efficacious in patients with higher DPP-4 expression in blocking IL-1b could be beneficial for preventing obesity-
adipose tissue. Importantly, recent data suggest that the associated insulin resistance and inflammation in human
role of DPP-4 in diabetes is not restricted to its incretin- adipose tissue [101,102]. Indeed, in the context of a con-
degrading properties. Under physiologic concentrations, trolled clinical trial including 70 patients with T2DM,
DPP-4 directly induced insulin resistance in adipocytes treatment with a recombinant human IL-1R antagonist
and skeletal muscle [88]. In mice, production of DPP-4 in (anakinra) was shown to improve glycemia and b cell
adipocytes was shown to be negatively regulated by high function [100]. In humans with obesity and diabetes, IL-
glucose concentration under physiological conditions, an 1b release increased with glycemic deterioration and de-
effect that was abolished in mouse models of diabetes creased after gastric bypass surgery in a recent study
[90]. In humans, DPP-4 is more highly expressed in adi- [103]. Moreover, in these human intervention studies,
pose tissue dysfunction [89], but recent data suggest that, IL-1b and the T cell cytokine IL-22 were identified as
in addition to increased expression, hypoxia-mediated key players in a paracrine inflammatory pathway in adi-
upregulation of DPP-4 shedding in adipocytes and smooth pose tissue [103]. However, the predominant beneficial
muscle cells by metalloproteases also contributes to higher effects of targeting IL-1b may be due to protection of
DPP-4 activity in obesity [91]. Therefore, DPP-4 may pancreatic b cells rather than improvement of peripheral
not only reflect, but also contribute to adipose tissue insulin sensitivity.
7
TIPS-1235; No. of Pages 10

Review Trends in Pharmacological Sciences xxx xxxx, Vol. xxx, No. x

Concluding remarks risks that a mechanistic concept derived from animal


Adipose tissue secretes hundreds of bioactive molecules, experiments does not translate into effective treatment
the majority of which are peptide hormones called adipo- in humans or that it causes inacceptable adverse effects.
kines. In obesity, metabolic and inflammatory diseases These important barriers have to be overcome in the next
adipokine secretion may change and thereby mechanisti- years to develop (more) adipokine-based pharmacothera-
cally link adipocyte hypertrophy and adipose tissue dys- pies (Box 1).
function in obesity to related metabolic, cardiovascular and
even malignant comorbidities. Adipokines are important Acknowledgments
modulators of appetite and satiety, energy expenditure This work was supported by a grant from Deutsche Forschungsge-
and activity, fat distribution, adipocyte function, glucose meinschaft (Sonderforschungsbereich 1052 ‘Obesity Mechanisms’; pro-
and lipid metabolism, insulin sensitivity, chronic inflam- jects B01 and C06), and by the Helmholtz Alliance ICEMED – Imaging
and Curing Environmental Metabolic Diseases, through the Initiative
mation, and other processes. Over recent years, many
and Networking Fund of the Helmholtz Association.
novel adipokines have been discovered. The main chal-
lenge in adipokine research remains to elucidate their
function and main targets. Understanding the mode of References
1 Van Gaal, L.F. et al. (2006) Mechanisms linking obesity with
action for novel adipokines may facilitate their future
cardiovascular disease. Nature 444, 875–880
clinical use as pharmacotherapies, drug targets, or pre- 2 LeRoith, D. et al. (2008) Obesity and type 2 diabetes are associated
dictors of a variety of diseases. with an increased risk of developing cancer and a worse prognosis;
In summary, adipokines are promising candidates for epidemiological and mechanistic evidence. Exp. Clin. Endocrinol.
the future treatment of obesity and obesity-related dis- Diabetes 116, S4–S6
3 Blüher, M. (2013) Importance of adipokines in glucose homeostasis.
eases. Over the past few years, drug discovery paths have Diabetes Manage. 3, 389–400
been advanced to clinical studies for the use of leptin, 4 Klöting, N. and Blüher, M. (2014) Adipocyte dysfunction,
FGF21 and BMP-7 as therapeutics, as well as DPP-4, inflammation and metabolic syndrome. Rev. Endocr. Metab. Disord.
TNFa, and IL-1b as targets for pharmacotherapy. For 15, 277–287
adiponectin and the majority of more recently discovered 5 Blüher, M. (2013) Adipose tissue dysfunction contributes to obesity
related metabolic diseases. Best Pract. Res. Clin. Endocrinol. Metab.
adipokines, we may expect efforts to translate promising 27, 163–177
data from animal studies into human clinical trials within 6 Kershaw, E.E. and Flier, J.S. (2004) Adipose tissue as an endocrine
the next 2–4 years. However, the drug development pro- organ. J. Clin. Endocrinol. Metab. 89, 2548–2556
cess may take approximately 15 years from the discovery of 7 Lehr, S. et al. (2012) Adipokines: a treasure trove for the discovery
of biomarkers for metabolic disorders. Proteomics Clin. Appl. 1–2,
a candidate adipokine to its clinical use or approval by
91–101
authorities. 8 Lehr, S. et al. (2012) Identification and validation of novel adipokines
For most adipokines, the major outstanding question is released from primary human adipocytes. Mol. Cell. Proteomics 11,
how altered secretion leads to metabolic, cardiovascular, http://dx.doi.org/10.1074/mcp.M111.010504 M111.010504
inflammatory, and malignant diseases. The field could be 9 Dahlman, I. et al. (2012) Functional annotation of the human fat cell
secretome. Arch. Physiol. Biochem. 118, 84–91
driven forward by the characterization of the mode of
10 Blüher, M. and Mantzoros, C.S. (2015) From leptin to other
action of adipokines and identification of molecular adipo- adipokines in health and disease: facts and expectations at the
kine targets. In the drug discovery path, there are always beginning of the 21st century. Metabolism 64, 131–145
11 Blüher, M. (2014) Adipokines: removing road blocks to obesity and
diabetes therapy. Mol. Metab. 3, 230–240
12 Blüher, M. (2012) Clinical relevance of adipokines. Diabetes Metab. J.
36, 317–327
Box 1. Outstanding questions 13 Björntorp, P. and Sjöström, L. (1971) Number and size of adipose
tissue fat cells in relation to metabolism in human obesity. Metab.
Questions Clin. Exp. 20, 703–713
 What are the unrecognized signals from adipose tissue that may 14 Cotillard, A. et al. (2014) Adipocyte size threshold matters: link with
promote overeating? risk of type 2 diabetes and improved insulin resistance after gastric
 Are there potential synergistic effects and concerted actions of bypass. J. Clin. Endocrinol. Metab. 99, E1466–E1470
adipokines (which will not be recognized by single gene deletion 15 Skurk, T. et al. (2007) Relationship between adipocyte size and
and/or overexpression studies in animals)? adipokine expression and secretion. J. Clin. Endocrinol. Metab. 92,
 Do adipokines have a role in food preferences? 1023–1033
 Do adipokines predict and/or promote weight regain following 16 Klöting, N. et al. (2010) Insulin-sensitive obesity. Am. J. Physiol.
intended weight loss? Endocrinol. Metab. 299, E506–E515
 Why are some individuals protected against the development of 17 Laurencikiene, J. et al. (2011) Regulation of lipolysis in small and
adverse adipokine secretion? large fat cells of the same subject. J. Clin. Endocrinol. Metab. 96,
 What are the molecular mechanisms linking altered adipokine E2045–E2049
secretion to metabolic, cardiovascular, inflammatory, and malig- 18 Schleinitz, D. et al. (2014) The genetics of fat distribution.
nant diseases? Diabetologia 57, 1276–1286
19 Zhang, Y. et al. (2014) Fat cell size and adipokine expression in
Unmet needs relation to gender, depot, and metabolic risk factors in morbidly
obese adolescents. Obesity 22, 691–697
 Definition of function, molecular action, and targets for the
20 Klöting, N. et al. (2007) Serum retinol-binding protein is more highly
majority of recently identified adipokines
expressed in visceral than in subcutaneous adipose tissue and is a
 Development of adipokine-based pharmacological treatment
marker of intra-abdominal fat mass. Cell Metab. 6, 79–87
strategies
21 Chakaroun, R. et al. (2012) Effects of weight loss and exercise on
 Establish specific adipokines as tools for the early diagnosis of
chemerin serum concentrations and adipose tissue expression in
diseases
human obesity. Metabolism 61, 706–714

8
TIPS-1235; No. of Pages 10

Review Trends in Pharmacological Sciences xxx xxxx, Vol. xxx, No. x

22 Youn, B.S. et al. (2009) Serum progranulin concentrations may be 50 Sook Lee, E. et al. (2013) Association between adiponectin levels and
associated with macrophage infiltration into omental adipose tissue. coronary heart disease and mortality: a systematic review and meta-
Diabetes 58, 627–636 analysis. Int. J. Epidemiol. 42, 1029–1039
23 Flehmig, G. et al. (2014) Identification of adipokine clusters related to 51 Spranger, J. et al. (2003) Adiponectin and protection against type
parameters of fat mass, insulin sensitivity and inflammation. PLoS 2 diabetes mellitus. Lancet 361, 226–228
ONE 9, e99785 52 Itoh, N. (2014) FGF21 as a hepatokine, adipokine, and myokine in
24 Cook, K.S. et al. (1987) Adipsin: a circulating serine protease homolog metabolism and diseases. Front. Endocrinol. 5, 107
secreted by adipose tissue and sciatic nerve. Science 237, 402–405 53 Gaich, G. et al. (2013) The effects of LY2405319, an FGF21 analog, in
25 Flier, J.S. et al. (1987) Severely impaired adipsin expression in genetic obese human subjects with type 2 diabetes. Cell Metab. 18, 333–340
and acquired obesity. Science 237, 405–408 54 Kharitonenkov, A. et al. (2005) FGF-21 as a novel metabolic regulator.
26 Lo, J.C. et al. (2014) Adipsin is an adipokine that improves b cell J. Clin. Invest. 115, 1627–1635
function in diabetes. Cell 158, 41–53 55 Fisher, F.M. et al. (2012) FGF21 regulates PGC-1a and browning
27 Siiteri, P.K. (1987) Adipose tissue as a source of hormones. Am. J. of white adipose tissues in adaptive thermogenesis. Genes Dev. 26,
Clin. Nutr. 45, 277–282 271–281
28 Zhang, Y. et al. (1994) Positional cloning of the mouse obese gene and 56 Cheng, X. et al. (2011) Serum FGF-21 levels in type 2 diabetic
its human homologue. Nature 372, 425–432 patients. Endocr. Res. 36, 142–148
29 Farooqi, I.S. et al. (1999) Effects of recombinant leptin therapy in a 57 Kharitonenkov, A. and Adams, A.C. (2014) Inventing new medicines:
child with congenital leptin deficiency. N. Engl. J. Med. 341, 879–884 the FGF21 story. Mol. Metab. 3, 221–229
30 Foo, J.P. and Mantzoros, C.S. (2012) Leptin in congenital or HIV- 58 Wei, W. (2012) Fibroblast growth factor 21 promotes bone loss by
associated lipodystrophy and metabolic syndrome: a need for more potentiating the effects of peroxisome proliferator-activated receptor
mechanistic studies and large, randomized, placebo-controlled trials. gamma. Proc. Nat. Acad. Sci. U.S.A. 109, 3143–3148
Metabolism 61, 1331–1336 59 Zhang, Y. (2012) The starvation hormone, fibroblast growth factor–21,
31 Mantzoros, C.S. et al. (2011) Leptin in human physiology and extends lifespan in mice. Elife 1, e00065
pathophysiology. Am. J. Physiol. Endocrinol. Metab. 301, E567–E584 60 Schulz, T.J. et al. (2013) Brown-fat paucity due to impaired BMP
32 Fasshauer, M. et al. (2014) Adipokines in gestational diabetes. Lancet signalling induces compensatory browning of white fat. Nature 495,
Diabetes Endocrinol. 2, 488–499 379–383
33 Heymsfield et al. (1999) Recombinant leptin for weight loss in obese 61 Schulz, T.J. and Tseng, Y.H. (2009) Emerging role of bone
and lean adults: a randomized, controlled, dose-escalation trial. morphogenetic proteins in adipogenesis and energy metabolism.
JAMA 282, 1568–1575 Cytokine Growth Factor Rev. 20, 523–531
34 Hukshorn, C.J. et al. (2000) Weekly subcutaneous pegylated 62 Tseng, Y.H. et al. (2008) New role of bone morphogenetic protein 7 in
recombinant native human leptin (PEG-OB) administration in brown adipogenesis and energy expenditure. Nature 454, 1000–1004
obese men. J. Clin. Endocrinol. Metab. 85, 4003–4009 63 Townsend, K.L. et al. (2012) Bone morphogenetic protein 7 (BMP7)
35 Roth, J.D. et al. (2008) Leptin responsiveness restored by amylin reverses obesity and regulates appetite through a central mTOR
agonism in diet-induced obesity: evidence from nonclinical and pathway. FASEB J. 26, 2187–2196
clinical studies. Proc. Natl. Acad. Sci. U.S.A. 105, 7257–7262 64 Vaccaro, A.R. et al. (2008) The safety and efficacy of OP-1 (rhBMP-7)
36 Ravussin, E. et al. (2009) Enhanced weight loss with pramlintide/ as a replacement for iliac crest autograft for posterolateral lumbar
metreleptin: an integrated neurohormonal approach to obesity arthrodesis: minimum 4-year follow-up of a pilot study. Spine J. 8,
pharmacotherapy. Obesity 17, 1736–1743 457–465
37 Moon, H.S. et al. (2013) Leptin’s role in lipodystrophic and 65 Gustafson, B. and Smith, U. (2012) The WNT inhibitor Dickkopf 1 and
nonlipodystrophic insulin-resistant and diabetic individuals. bone morphogenetic protein 4 rescue adipogenesis in hypertrophic
Endocr. Rev. 34, 377–412 obesity in humans. Diabetes 61, 1217–1224
38 Dalamaga, M. et al. (2013) Leptin at the intersection of 66 Gustafson, B. et al. (2015) BMP4 and BMP antagonists regulate
neuroendocrinology and metabolism: current evidence and human white and beige adipogenesis. Diabetes 64, 1670–1681
therapeutic perspectives. Cell Metab. 18, 29–42 67 Cypess, A.M. et al. (2009) Identification and importance of brown
39 Ye, R. and Scherer, P.E. (2013) Adiponectin, driver or passenger on adipose tissue in adult humans. N. Engl. J. Med. 360, 1509–1517
the road to insulin sensitivity? Mol. Metab. 2, 133–141 68 van Marken Lichtenbelt, W.D. et al. (2009) Cold-activated brown
40 Scherer, P.E. et al. (1995) A novel serum protein similar to C1q, adipose tissue in healthy men. N. Engl. J. Med. 360, 1500–1508
produced exclusively in adipocytes. J. Biol. Chem. 270, 26746–26749 69 Virtanen, K.A. et al. (2009) Functional brown adipose tissue in
41 Hu, E. et al. (1996) AdipoQ is a novel adipose-specific gene healthy adults. N. Engl. J. Med. 360, 1518–1525
dysregulated in obesity. J. Biol. Chem. 271, 10697–10703 70 Hida, K. et al. (2005) Visceral adipose tissue-derived serine protease
42 Maeda, K. et al. (1996) cDNA cloning and expression of a novel adipose inhibitor: a unique insulin-sensitizing adipocytokine in obesity. Proc.
specific collagen-like factor, apM1 (AdiPose Most abundant Gene Natl. Acad. Sci. U.S.A. 102, 10610–10615
transcript 1). Biochem. Biophys. Res. Commun. 221, 286–289 71 Youn, B.S. et al. (2008) Serum vaspin concentrations in human obesity
43 Turer, A.T. and Scherer, P.E. (2012) Adiponectin: mechanistic and type 2 diabetes. Diabetes 57, 372–377
insights and clinical implications. Diabetologia 55, 2319–2326 72 Blüher, M. (2012) Vaspin in obesity and diabetes: pathophysiological
44 Okamoto, M. et al. (2008) Adiponectin induces insulin secretion in vitro and clinical significance. Endocrine 41, 176–182
and in vivo at a low glucose concentration. Diabetologia 51, 827–835 73 Klöting, N. et al. (2011) Central vaspin administration acutely
45 Okada-Iwabu, M. et al. (2013) A small-molecule AdipoR agonist for reduces food intake and has sustained blood glucose-lowering
type 2 diabetes and short life in obesity. Nature 503, 493–499 effects. Diabetologia 54, 1819–1823
46 Kadowaki, T. et al. (2014) Adiponectin and its receptors: implications 74 Heiker, J.T. et al. (2013) Vaspin inhibits kallikrein 7 by serpin
for obesity-associated diseases and longevity. Lancet Diabetes mechanism. Cell. Mol. Life Sci. 70, 2569–2583
Endocrinol. 2, 8–9 75 Boucher, J. et al. (2005) Apelin, a newly identified adipokine up-
47 Wu, Z.J. et al. (2014) Adiponectin is associated with increased regulated by insulin and obesity. Endocrinology 146, 1764–1771
mortality in patients with already established cardiovascular 76 Castan-Laurell, I. et al. (2011) Apelin, diabetes, and obesity.
disease: a systematic review and meta-analysis. Metabolism 63, Endocrine 40, 1–9
1157–1166 77 Kuba, K. et al. (2007) Impaired heart contractility in Apelin gene-
48 Karas, M.G. et al. (2014) Relations of plasma total and high- deficient mice associated with aging and pressure overload. Circ. Res.
molecular-weight adiponectin to new-onset heart failure in adults 101, e32–e42
>/=65 years of age (from the Cardiovascular Health study). Am. J. 78 Yue, P. et al. (2011) Apelin decreases lipolysis via G(q), G(i), and
Cardiol. 113, 328–334 AMPK-dependent mechanisms. Endocrinology 152, 59–68
49 Cote, M. et al. (2011) Adiponectin and risk of coronary heart disease in 79 Cao, J. et al. (2015) Targeting drugs to APJ receptor: the prospect of
apparently healthy men and women (from the EPIC-Norfolk treatment of hypertension and other cardiovascular diseases. Curr.
Prospective Population Study). Am. J. Cardiol. 108, 367–373 Drug Targets 16, 148–155

9
TIPS-1235; No. of Pages 10

Review Trends in Pharmacological Sciences xxx xxxx, Vol. xxx, No. x

80 Frühbeck, G. et al. (2014) Regulation of adipocyte lipolysis. Nutr. Res. 92 Dunmore, S.J. and Brown, J.E.P. (2013) The role of adipokines in b-
Rev. 27, 63–93 cell failure of type 2 diabetes. J. Endocrinol. 216, T37–T45
81 Krist, J. et al. (2013) Effects of weight loss and exercise on apelin 93 Cawthorn, W.P. and Sethi, J.K. (2008) TNF-alpha and adipocyte
serum concentrations and adipose tissue expression in human biology. FEBS Lett. 582, 117–131
obesity. Obes. Facts 6, 57–69 94 Hotamisligil, G.S. et al. (1993) Adipose expression of tumor necrosis
82 Sorhede Winzell, M. et al. (2005) The apj receptor is expressed in factor-alpha: direct role in obesity-linked insulin resistance. Science
pancreatic islets and its ligand, apelin, inhibits insulin secretion in 259, 87–91
mice. Regul. Pept. 131, 12–17 95 Uysal, K.T. et al. (1997) Protection from obesity-induced insulin
83 Dray, C. et al. (2008) Apelin stimulates glucose utilization in normal resistance in mice lacking TNF-alpha function. Nature 389, 610–614
and obese insulin-resistant mice. Cell Metab. 8, 437–445 96 Li, Z. et al. (2003) Probiotics and antibodies to TNF inhibit
84 Campbell, J.E. and Drucker, D.J. (2013) Pharmacology, physiology, inflammatory activity and improve nonalcoholic fatty liver disease.
and mechanisms of incretin hormone action. Cell Metab. 17, 819–837 Hepatology 37, 343–350
85 Nauck, M.A. (2011) Incretin-based therapies for type 2 diabetes 97 Liang, H. et al. (2008) Blockade of tumor necrosis factor (TNF)
mellitus: properties, functions, and clinical implications. Am. J. receptor type 1-mediated TNF-alpha signaling protected Wistar
Med. 124, S3–S18 rats from diet-induced obesity and insulin resistance.
86 Drucker, D.J. (2013) Incretin action in the pancreas: potential Endocrinology 149, 2943–2951
promise, possible perils, and pathological pitfalls. Diabetes 62, 98 Hmamouchi, I. et al. (2014) Early increase of abdominal adiposity in
3316–3323 patients with spondyloarthritis receiving anti-tumor necrosis factor-a
87 Mulvihill, E.E. and Drucker, D.J. (2014) Pharmacology, physiology, treatment. J. Rheumatol. 41, 1112–1117
and mechanisms of action of dipeptidyl peptidase-4 inhibitors. 99 Wascher, T.C. et al. (2011) Chronic TNF-alpha neutralization does not
Endocr. Rev. 35, 992–1019 improve insulin resistance or endothelial function in ‘healthy’ men
88 Lamers, D. et al. (2011) Dipeptidyl peptidase 4 is a novel adipokine with metabolic syndrome. Mol. Med. 17, 189–193
potentially linking obesity to the metabolic syndrome. Diabetes 60, 100 Larsen, C.M. et al. (2007) Interleukin-1-receptor antagonist in type
1917–1925 2 diabetes mellitus. N. Engl. J. Med. 356, 1517–1526
89 Sell, H. et al. (2013) Adipose dipeptidyl peptidase-4 and obesity: 101 Tack, C.J. et al. (2012) Inflammation links excess fat to insulin
correlation with insulin resistance and depot-specific release from resistance: the role of the interleukin–1 family. Immunol. Rev. 249,
adipose tissue in vivo and in vitro. Diabetes Care 36, 4083–4090 239–252
90 Das, S.S. et al. (2014) Regulation of dipeptidyl peptidase 4 production 102 Gao, D. (2014) Interleukin-1b mediates macrophage-induced
in adipocytes by glucose. Diabetes Metab. Syndr. Obes. 7, 185–194 impairment of insulin signaling in human primary adipocytes. Am.
91 Röhrborn, D. et al. (2014) Shedding of dipeptidyl peptidase 4 is J. Physiol. Endocrinol. Metab. 307, E289–E304
mediated by metalloproteases and up-regulated by hypoxia in 103 Dalmas, E. (2014) T cell-derived IL-22 amplifies IL-1b-driven
human adipocytes and smooth muscle cells. FEBS Lett. 588, inflammation in human adipose tissue: relevance to obesity and
3870–3877 type 2 diabetes. Diabetes 63, 1966–1977

10

Vous aimerez peut-être aussi