You are on page 1of 18

REVIEWS

The endocrine function of adipose


tissues in health and cardiometabolic
disease
Ludger Scheja and Joerg Heeren *
Abstract | In addition to their role in glucose and lipid metabolism, adipocytes respond
differentially to physiological cues or metabolic stress by releasing endocrine factors that
regulate diverse processes, such as energy expenditure, appetite control, glucose homeostasis,
insulin sensitivity, inflammation and tissue repair. Both energy-storing white adipocytes and
thermogenic brown and beige adipocytes secrete hormones, which can be peptides (adipokines),
lipids (lipokines) and exosomal microRNAs. Some of these factors have defined targets;
for example, adiponectin and leptin signal through their respective receptors that are expressed
in multiple organs. For other adipocyte hormones, receptors are more promiscuous or remain to
be identified. Furthermore, many of these hormones are also produced by other organs and
tissues, which makes defining the endocrine contribution of adipose tissues a challenge. In this
Review, we discuss the functional role of adipose tissue-derived endocrine hormones for
metabolic adaptations to the environment and we highlight how these factors contribute to the
development of cardiometabolic diseases. We also cover how this knowledge can be translated
into human therapies. In addition, we discuss recent findings that emphasize the endocrine role
of white versus thermogenic adipocytes in conditions of health and disease.

Leptin, which is a peptide hormone expressed by adi- hormones, which are released from adipose tissues and
pocytes that is essential for body weight control and exert defined responses in target organs. Nevertheless,
many other functions, was discovered in 1994. Its dis- many studies indicate endocrine roles for various other
covery established adipose tissue as an essential endo- factors. Overall, unbiased approaches have found a very
crine organ1. Since then, research has revealed many large number of molecules with proven or potential sig-
adipose tissue-derived peptide hormones (adipokines) nalling activity that are secreted by adipocytes, which are
and non-peptide endocrine factors, which are the topic lipid-storing connective tissue cells found throughout
of this Review (Box 1; Fig. 1). Important highlighted the body, and other adipose tissue cells3. For example, a
aspects include regulation of the expression and secre- 2018 proteomic study compared the secretome of mouse
tion of adipose tissue endocrine factors as well as their white and brown adipocytes and identified more than
modes of action. We focus on the impact of the acti- 1000 unique secreted proteins, of which more than 100
vation state of adipose tissue depots for the secretion were differentially released between white and brown
of endocrine factors. Moreover, we discuss the role of adipocytes4. Another comprehensive bioinformatics
brown adipose tissue (BAT) compared with that of the study, based on RNA sequencing data from more than
various white adipose tissue (WAT) depots. In addition, 100 different mouse strains, addressed endocrine inter-
we present the current views of how adipose tissue hor- actions between adipose tissue and various organs.
mones contribute to the development and progression of Using this elegant approach, researchers identified sev-
cardiometabolic diseases in animal models and humans, eral novel adipose tissue-derived endocrine candidates,
Department of Biochemistry including therapeutic implications. such as lipocalin 5 (ref.5).
and Molecular Cell Biology, Notably, only a few of the factors released by adi- Owing to the sheer number of potential adipose
University Medical Center
Hamburg-Eppendorf,
pose tissue are established endocrine hormones, and tissue-derived endocrine factors, we can cover only a
Hamburg, Germany. many of them, especially those molecules secreted by small number in this Review. We focus on hormones
*e-mail: heeren@uke.de non-adipocytes, exert predominantly paracrine func- where endocrine functions have been most compre-
https://doi.org/10.1038/ tions2. Only leptin and adiponectin are currently gene­ hensively studied and on recently identified candidates.
s41574-019-0230-6 rally accepted as true adipose tissue-derived endocrine Furthermore, factors were selected in a way to represent

NATuRe RevIeWS | EndOcRInOlOgy volume 15 | SEPTEMBER 2019 | 507


Reviews

Key points adipocyte hypertrophy and fibrosis10, local inflammation,


infiltration of immune cells, polarization of macrophages
• White and brown adipocytes secrete many peptide hormones (adipokines), bioactive from an anti-inflammatory to a pro-inflammatory pheno­
lipids (lipokines) and RNA molecules with local (paracrine) and systemic (endocrine) type (Fig. 2), altered lipid handling in macrophages,
effects on the brain, pancreatic β-cells, the liver, skeletal muscle and the decreased response of adipocytes to insulin (insulin
cardiovascular system.
resistance) and a disturbed metabolism11–14.
• Production and secretion of adipokines and lipokines is dependent on the energy Adipocyte insulin resistance leads to insufficient lipid
status of adipose tissues. Through endocrine action, these factors contribute to
retention in WAT and, consequently, ectopic lipid accu-
systemic energy metabolism by regulating appetite, thermogenesis, glucose
metabolism and lipid metabolism.
mulation occurs in organs such as the liver and skeletal
muscle. This accumulation is hypothesized to disturb
• Many peptides that were initially described as adipokines are secreted by endothelial
and immune cells located in adipose tissues, as well as by other organs, which means
insulin signalling in those organs and further promote
the endocrine contribution of adipocytes can be difficult to ascertain. the development of systemic insulin resistance and type 2
• In healthy states, white and brown adipose tissues secrete endocrine factors that
diabetes mellitus (T2DM)15 (Box 2). Importantly, fatty
maintain organ functions and metabolic homeostasis. acids released from white adipocytes, especially satu­rated
• In obesity, hypertrophic adipocytes and adipose tissue-resident immune cells
fatty acids such as palmitic acid, alter the production
accelerate a chronic, proinflammatory profile with altered secretion of adipokines and secretion of inflammatory cytokines and adi-
and lipokines, thereby exacerbating cardiometabolic disease. pokines from hypertrophic WAT, thereby contributing
• Preclinical and clinical studies show that activating or inhibiting the signalling of to obesity-related organ dysfunction (see later)8,10,11.
specific adipokines or lipokines could be an approach suitable to treat or prevent the Of note, not all WAT depots are associated with equal
development of cardiometabolic diseases. However, in almost all cases, efficacy and disease risk in human obesity. The amount of visceral
safety in humans needs to be proven. abdominal WAT, as compared with subcutaneous WAT,
is tightly linked with insulin resistance and metabolic
disease in humans7,16,17. The reason for this association is
the diverse primary biological functions of these mole- not fully clear but might be partially due to the drainage
cules beyond classical peptide hormone signalling. For of visceral fat into the portal vein7. This anatomical situ-
each molecule, we critically discuss the level of experi- ation means that molecules released from visceral WAT
mental evidence, important differences between rodents can reach the liver in higher concentrations than mole­
and humans (if they occur) and the gaps in knowledge cules derived from subcutaneous WAT7. Importantly,
identifying the molecule as a true endocrine factor. unhealthy obesity is associated with intra-abdominal
expansion of insulin-resistant and inflamed WAT16. As
The diversity of adipose tissue depots described later, some adipokines are enriched in visceral
White adipose tissue WAT and probably have a particular impact on the liver
WAT is the main site of energy storage in the body and in the context of visceral obesity.
is present in multiple anatomical locations6,7. White
adipo­cytes are the predominate cell type found in WAT, Brown adipose tissue
and contain low amounts of mitochondria and none of BAT is a highly vascularized organ rich in brown adipo­
these mitochondria contain uncoupling protein 1 (UCP1). cytes, which is unique to mammals and functions to gen-
In the postprandial state (that is, after a meal), white erate heat by metabolizing fatty acids and glucose in a
adipo­cytes take up fatty acids carried by circulating lipo­ process called ‘adaptive (non-shivering) thermogenesis’.
proteins, convert them into triglycerides and store these In contrast to white adipocytes, brown adipocytes have
in a large intracellular lipid droplet. By contrast, between multiple small lipid droplets as well as a much higher
meals and in other catabolic states, WAT releases free content of mitochondria, and, importantly, these mito-
fatty acids (FFAs) liberated by intracellular lipolysis into chondria contain UCP1, which shuttles protons back
the bloodstream to provide other organs with energy8. into the matrix and uncouples the electron transfer chain
In adipocytes, catecholamines, which originate primar- from ATP synthesis, thereby generating heat18. The pro-
ily from the sympathetic nervous system, are the major cess of adaptive thermogenesis is triggered by increased
lipolytic signalling molecules and increase intracellular sympathetic tone or by the action of endocrine hor-
cyclic AMP levels by stimulating β-adrenergic receptors. mones such as natriuretic peptides and secretin9,19, which
Moreover, other hormones, such as natriuretic peptides9, stimulate intracellular lipolysis in brown adipocytes.
induce catabolic signalling by elevating intracellular
cyclic AMP or cyclic GMP concentrations. By contrast,
in the postprandial state, insulin inhibits lipolysis and Box 1 | Adipokines and lipokines
causes the uptake of fatty acids and glucose into WAT. In addition to their prominent role in fatty acid and
Not surprisingly, several endocrine factors released from energy metabolism, adipocytes and other cells in adipose
adipose tissues are regulated by insulin or sympathetic tissue secrete a large number of different regulatory
Uncoupling protein 1 tone, as described in detail later. peptides (adipokines)8,30,31, as well as regulatory lipid
(UCP1). A unique proton During periods of calorie surplus, WAT mass expands species (lipokines)8,90,97,104. These factors can act on
transporter that shuttles by increasing both the size (hypertrophy) and the number neighbouring cells (paracrine action) or on cells in
protons back into the of adipocytes (hyperplasia). However, chronic activation of other organs (endocrine action) to specifically regulate
mitochondrial matrix and or modulate diverse processes, including lipid and
uncouples the electron transfer
this process becomes detrimental, leading to obesity. In
obesity, excessive WAT expansion is accompanied by glucose homeostasis, energy balance, inflammation
chain from ATP synthesis,
and tissue repair.
a process that generates heat. structural and cellular changes in the tissue, including

508 | SEPTEMBER 2019 | volume 15 www.nature.com/nrendo


Reviews

WAT Brain
• Leptin • Adiponectin Leptin • Appetite
• Adiponectin • FABP4 • Temperature
• Adipsin • FAHFAs • Glucose homeostasis
• FABP4 • Palmitoleate
• FAHFAs
• Palmitoleate
Pancreas (β-cells)
• Leptin • Insulin secretion
• Adiponectin • β-Cell survival
• Adipsin
• FAHFAs

Cardiovascular system
Beige WAT
• Leptin • Inflammation
• Adiponectin • Vascular remodelling
• NRG4 • FABP4 • Vascular tone
• NRG4 • 12,13-diHOME • Palmitoleate
• 12,13-diHOME

Liver
• Adiponectin • Glucose metabolism
• FABP4 • Lipid metabolism
• Palmitoleate • Insulin signalling
• NRG4
• 12,13-diHOME
• miR-99b
BAT

• NRG4 Musculature
• 12,13-diHOME • NRG4
• miR-99b • 12,13-diHOME • Adiponectin • Glucose metabolism
• miR-92a • miR-92a • Palmitoleate • Lipid metabolism
12,13-diHOME • Insulin signalling

Fig. 1 | Endocrine effects of adipocyte-secreted factors. Adipocyte-secreted hormones have multiple effects on other
organs. Although most adipose tissue hormones are produced by both white adipose tissue (WAT) and brown adipose tissue
(BAT), only major sites of origin are shown. Adiponectin is downregulated in obesity and is important for maintaining glucose
homeostasis, insulin sensitivity and organ function in chronic inflammatory states. The circulating levels of leptin, which
suppresses appetite, improves peripheral insulin resistance, raises body temperature and regulates hormonal axes, correlate
with total adipose tissue mass. The protease adipsin is an essential component of the alternative complement pathway and
stimulates insulin secretion via C3a receptors on β-cells. In obesity, increased plasma levels of the lipid chaperone fatty
acid-binding protein 4 (FABP4) increase hepatic gluconeogenesis, peripheral insulin resistance and plaque formation in the
arteries. By contrast, levels of the anti-inflammatory fatty acid palmitoleate are decreased in obesity. Fatty acid esters of
hydroxy fatty acids (FAHFAs) are a novel class of lipids that improve glucose tolerance by increasing adipocyte glucose
transport and insulin secretion. Exosomal microRNA (miR)-99b might suppress hepatic fibroblast growth factor 21 production,
whereas miR-92a serves as a biomarker for BAT. Production of the fatty acid derivative 12,13-dihydroxy-(9Z)-octadecenoic
acid (12,13-diHOME) is increased in thermogenic adipose tissues in response to cold exposure to facilitate thermogenesis.
Neuregulin 4 (NRG4) is released from activated thermogenic adipose tissue, acting on hepatocytes to decrease de novo
lipogenesis and prevent liver damage. Dashed arrows indicate paracrine activity; solid arrows indicate endocrine activity.

Subsequently, fatty acids that are released from lipid substantial metabolic potential21. Superimposed on the
droplets can allosterically activate UCP1 and are then effects of sympathetic tone, other mechanisms have been
combusted in the mitochondria to generate heat18. demonstrated to regulate the abundance and function of
In parallel with BAT activation, the replenishment of brown and beige adipocytes. For example, innate immune
energy stores in BAT is initiated, which is a process cells such as tissue-resident M2 macrophages have been
regulated by both insulin and β-adrenergic signalling20. shown to induce thermogenesis in both BAT and WAT,
Under prolonged stimulation, BAT adapts by increas- although the mechanisms remain controversial22.
ing the mass and the number of brown adipocytes18 The abundance of BAT differs substantially in human
(Fig. 2). Of note, cold exposure or other catabolic condi- populations and typically decreases with advanced age
M2 macrophages
Macrophages polarized by tions can trigger an adaptive process in WAT in rodents and increasing body weight, thereby suggesting a role in
type 2 immunity-related and humans, which occurs when WAT is continuously age-related and obesity-related diseases21. In lean adults,
cytokines such as IL-4 and stimulated by the sympathetic nervous system 18,21. BAT is present at distinct locations in the upper body,
IL-13, which maintain tissue Subsequently, UCP1-containing adipocytes (so-called predominantly in thyroid, supraclavicular, mediastinal,
homeostasis by counteracting
proinflammatory processes
beige (or brite) adipocytes) appear among white adipo­ parathoracic and perirenal depots6. In contrast to mice,
and facilitating tissue cytes; these beige adipocytes are functionally very sim- human BAT depots are relatively small. Nevertheless,
regeneration. ilar or even identical to brown adipocytes and have owing to the exceptionally high metabolic activity of

NATuRe RevIeWS | EndOcRInOlOgy volume 15 | SEPTEMBER 2019 | 509


Reviews

WAT BAT

• Inactivity • Exercise
• Thermoneutrality • Cold exposure
• Caloric excess • Caloric restriction

Hypertrophic WAT and whitened BAT Healthy WAT and active BAT
Saturated FFAs ↑ ↓
Anorexigenic peptide ↑ Leptin ↓ Leptin
Anti-inflammatory lipids ↓ Palmitoleate, FAHFAs ?
Anti-inflammatory peptides ↓ Adiponectin, omentin, NRG4 ↑ Adiponectin, NRG4
Proinflammatory peptides ↑ RBP4, FABP4, resistin, IL-6 ↑ IL-6
Prothermogenic molecules ↓ CXCL14 ↑ CXCL14, 12,13-diHOME

Proinflammatory tissue-resident immune cell Anti-inflammatory tissue-resident immune cell

Fig. 2 | Endocrine factors released by healthy and unhealthy adipose tissues. In response to chronic energy excess,
an infiltration of proinflammatory immune cells can be observed in adipose tissue, which develops a hypertrophic white
adipose tissue (WAT) and whitened brown adipose tissue (BAT) phenotype, a hallmark of unhealthy obesity. In contrast,
energy expenditure or decreased food intake results in healthy WAT and BAT, which is characterized by the presence of
anti-inflammatory immune cells and by active thermogenic brown and beige adipocytes. Both healthy and metabolic
disease states are accompanied by altered secretion of adipose tissue hormones as indicated by the arrows. Overall, the
changes in hypertrophic WAT contribute to insulin resistance, impaired glucose and lipid metabolism and low-grade
systemic inflammation. In catabolic states, thermogenic adipocytes secrete various endocrine factors that ensure
metabolic homeostasis and tissue remodelling. CXCL14, C-X-C motif chemokine ligand 14; 12,13-diHOME, 12,13-­dihydroxy-
(9Z)-octadecenoic acid; FABP4, fatty acid-binding protein 4; FAHFAs, fatty acid esters of hydroxy fatty acids; FFAs, free
fatty acids; NRG4, neuregulin 4; RBP4, retinol-binding protein 4.

BAT, these small amounts of tissue still have an impact Arteries and arterioles are surrounded by adipocytes
on whole body metabolism21. BAT also produces a num- that share features of brown and beige adipocytes, which
ber of endocrine factors (Figs. 1,2). Although the much comprise the perivascular adipose tissue (PVAT). These
larger WAT depots are probably the dominant source cells are involved in the regulation of vasodilation, pro-
of most circulating adipose tissue factors, especially vide mechanical support to the vessel, buffer excessive
in humans, some hormones are selectively induced in fatty acids and release paracrine hormones26. Of note,
BAT under certain conditions and probably act in an increased inflammation in PVAT is associated with higher
endocrine fashion, as described later. cardiovascular disease risk in humans and increases the
development of atherosclerosis in mice26–28. Analogous to
Organ-associated adipose tissue depots PVAT, a layer of adipose tissue covers the myocardium and
Some specialized adipose tissue depots are closely asso- the coronary arteries of the heart. This epicardial adipose
ciated with other organs and may affect these organs tissue (ECAT) is functionally similar to PVAT and shares
by contributing to the tissue microenvironment. One the microcirculation with cardiomyocytes. Consequently,
example is bone marrow adipose tissue (BMAT), which ECAT can have a negative impact on the myocardium in
is particularly abundant in the medulla of long bones. The states of metabolic inflammation29. In conclusion, local
data available suggest that bone marrow adipocytes have adipocytes are closely connected with the cells constitut-
primarily local functions such as the regulation of bone ing the cardiovascular system, conferring both protection
turnover23 and haematopoiesis24. In addition, bone mar- from disease and increased risk of disease.
row adipocytes might also have systemic effects through Of note, most studies investigating adipose tissue
secretion of the adipokine adiponectin25 (see later). biology focus on classical WAT and BAT depots. As such,

510 | SEPTEMBER 2019 | volume 15 www.nature.com/nrendo


Reviews

Box 2 | Unhealthy adipose tissues role in metabolism, leptin has substantial effects on the
hypothalamus–pituitary hormonal axis, the regulation of
In obesity, adipose tissues can become unhealthy when fertility, the immune system and bone turnover31.
adipocytes expand owing to chronic energy excess. In healthy conditions in humans and rodents, cir-
In this state, adipose tissues contain large (hypertrophic)
culating leptin levels correlate positively with adipose
adipocytes that are insulin-resistant, lose their ability to
tissue mass and leptin levels decrease sharply during pro-
adequately store triglycerides and display impaired
energy expenditure8,10,15. Consequently, fatty acids are longed fasting34, but do not substantially change between
released into the circulation and accumulate in other meals35. Thus, low plasma leptin levels can be regarded
organs, causing cellular stress and disturbed metabolism, as an endocrine signal reflecting depleted adipose tissue
which is a hallmark of chronic metabolic diseases such as energy stores and high energy demand. In line with this
type 2 diabetes mellitus, cardiovascular disease and notion, low plasma leptin concentration not only fosters
nonalcoholic steatohepatitis8,10,11,15. In addition, the increased energy intake but is also causally involved in
altered secretion of endocrine factors from hypertrophic starvation-associated alterations such as increased corti-
adipose tissues contributes to pathological changes in costerone and decreased thyroid hormone levels, gonadal
other organs8,30.
hypoactivity and suppression of the immune system36.
Leptin is expressed in all types of adipose tissues, with
the relevance of PVAT, ECAT and BMAT for endocrine a preference for subcutaneous WAT in humans37, and is
hormonal regulation requires further investigation. predominantly regulated at the transcriptional level38.
A general drawback of studying specific WAT depots is the In the fasted state, the sympathetic nervous system act-
lack of genetic intervention tools, for example, mouse ing on adipocyte β-adrenergic receptors is the principal
lines deficient for a gene of interest in specific adipose leptin-lowering mechanism39–41. By this mechanism, the
tissue depots. Currently, depot-specific in vivo research secretion of leptin is tightly coupled to nutritional state
is based on cumbersome surgical procedures such as and thus can mediate physiological adaptations.
excision and transplantation. For those WAT depots, a LEPR is a type I cytokine receptor that is expressed
more extensive use of local genetic manipulation (for in many cell types both in the central nervous sys-
example, using adeno-associated virus vectors) might be tem and in the periphery. In the brain, leptin signal-
useful to decipher the functional role of WAT as source ling is important for mediating the metabolic effects
of paracrine or endocrine factors. of leptin, as demonstrated by elevated levels of body
fat, glucose and insulin in neuron-specific but not in
Adipocyte-enriched endocrine factors hepatocyte-specific LEPR-deficient mice42. Evidence
Numerous regulatory factors secreted from adipose tis- is accumulating that all the major metabolic effects of
sue with impact on adjacent or remote tissues have been leptin are linked to leptin signalling in specific neurons
identified30 (Fig. 1; Tables 1,2) and discovery is ongoing5. or areas of the brain, resulting in increased sympathetic
Notably, of these factors only leptin and adiponectin flow to adipose tissues and subsequent higher energy
are selectively expressed in adipocytes and also well expenditure39,43, decreased heat loss32 and the suppres-
established as endocrine hormones that act on specific sion of appetite44,45, as well as the reduction of hepatic
receptors in remote target organs. In this section we gluconeogenesis and the prevention of peripheral insulin
describe these two classical adipokines, as well as other resistance46,47. Of note, two distinct neuron classes in the
adipokines, lipid hormones and regulatory RNAs that arcuate nucleus of the hypothalamus have been identi-
are predominantly produced by adipocytes or adipose fied as key leptin-responsive neurons: the agouti-related
tissues and that exert a defined endocrine function. For protein neurons and the pro-opiomelanocortin neurons.
each factor, we discuss the level of evidence indicating Selective knockdown of LEPR in either of these neuron
that it is part of a bona fide endocrine axis. classes or re-expression on a receptor-deficient back-
ground had substantial metabolic effects47,48. The meta­
Leptin and adiponectin bolic phenotype induced by such selective knockdown
Leptin. The obesity gene (ob) encodes an adipokine, is not as pronounced as that associated with systemic
called leptin, which is almost exclusively expressed LEPR knockdown, which suggests the importance
in adipocytes1 (Table 1). Leptin-deficient ob/ob mice of LEPR expressed in other parts of the brain (for example,
have an extremely obese phenotype, which is currently the hindbrain)44.
explained by the essential role of leptin in suppressing Notably, LEPR signalling in the periphery is also
appetite and promoting energy expenditure through relevant. For example, leptin could be antiatherogenic,
receptors in the central nervous system31; however, the as indicated by increased neointima formation in an
latter finding was challenged by a 2017 study demon- endothelial cell-specific Lepr-knockout mouse model49.
Lipodystrophy strating that leptin regulates heat loss rather than energy Leptin signalling also occurs in pancreatic β-cells50,51.
A genetic or acquired condition
expenditure32. Importantly, rodents or humans that lack In vitro studies found that leptin signalling inhibits
that is characterized by lack of
adipose tissue, insulin either leptin or the leptin receptor (LEPR) are not only insulin secretion and increases the survival of β-cells50,51.
resistance and hyperglycaemia. very obese but are also hyperglycaemic and extremely However, a 2015 study using a mouse model with LEPR
insulin-resistant31,33. These metabolic derangements knockdown restricted to β-cells found no evidence of
Neointima are not caused by excessive adipose tissue mass but hyperinsulinaemia or disturbed glucose homeostasis52.
Pathological thickening of the
subendothelial layer (intima) of
are due to the lack of leptin, as hyperglycaemia and Thus, leptin signalling in β-cells affects insulin secretion
arteries due to atherosclerosis insulin resistance are also present in lipodystrophy, a in vitro, but is seemingly not essential for maintaining
or other arterial injuries condition of very low leptin levels31. In addition to its functionality of β-cells in vivo. Overall, leptin signalling

NATuRe RevIeWS | EndOcRInOlOgy volume 15 | SEPTEMBER 2019 | 511


Reviews

Table 1 | Major cellular sources of adipose tissue-derived hormones


Factor White Brown and beige Preadipocytes Macrophages Endothelial cells Immune Hepatocytes Other
adipocytes adipocytes cells cells
Adipocyte-enriched endocrine factors
Leptin1,38 ++ ++ – – – – – –
Adiponectin25,53 ++ ++ – – – – – –
Adipsin 72
++ ++ – – – – – –
FABP4 (refs78,79) ++ ++ – ++ ++ – – –
NRG4 (refs 83,84
) – ++ – – – – ++ –
Lipokines
Palmitoleate90 ++ – – – – – – –
DNL FAHFAs97,98 ++ – – – – – – –
12,13-diHOME 104
– ++ – – – – – –
Adipocyte-derived exosomal microRNAs
Exosomal miR-99b108 ++ ++ – – – – – –
Exosomal miR-92b107 – ++ – – – – – –
Adipokines also released by hepatocytes
FGF21 (refs113,114,120,122) ++ ++, + – – – – ++ –
RBP4 (refs 30,129
) ++ ++ – – – – ++ –
IGF1 (ref. ) 197
++ ++, + – ++ – – ++ –
Adipokines also released by immune cells
CXCL14 (refs133,134) – ++ – – ++ ++ – ++
IL-6 (refs136,198) ++ ++ – ++ ++ ++ – –
Resistin 146,149
++ ++ – ++ – – – –
Broadly expressed adipokines
GDF15 (refs151,153,157) + + + + – + + +
eNAMPT159 ++ ++ ++ ++ – ++ ++ ++
Omentin 164,165
– – – ++ ++ – – ++
Vaspin169,199 ++ ++ ++ – – – – ++
+, factor is released from adipocytes under certain conditions; ++, factor is released from cell type; –, factor is not released in major amounts from cell type or
unknown; 12,13-diHOME, 12,13-dihydroxy-(9Z)-octadecenoic acid; CXCL14, C-X-C motif chemokine ligand 14; DNL , de novo lipogenesis; eNAMPT, extracellular
nicotinamide phosphoribosyltransferase; FABP4, fatty acid-binding protein 4; FAHFAs, fatty acid esters of hydroxy fatty acids; FGF21, fibroblast growth factor 21;
GDF15, growth differentiation factor 15; IGF1, insulin-like growth factor 1; NRG4, neuregulin 4; RBP4, retinol-binding protein 4.

in target cells outside the central nervous system has not high in bone marrow adipocytes of rabbits and mice
been studied as extensively, and more in vivo research is and that under specific conditions, such as calo-
required to dissect the role of this pleiotropic hormone rie restriction, expanded BMAT mass could explain
in physiology and pathophysiology. increased plasma adiponectin levels as compared with
ad libitum-fed controls25,57.
Adiponectin. All adipose tissue depots express adi- The action of adiponectin is important for systemic
ponectin, which is an adipocyte-specific hormone with insulin sensitivity, as demonstrated by the development
systemic insulin-sensitizing and anti-inflammatory of insulin resistance in adiponectin-deficient mice58
properties53 (Fig. 1). Plasma levels of this adipokine and by preserved insulin sensitivity in obese mice over­
are decreased in obese rodents and humans, and expressing adiponectin59. Adiponectin signalling is initi­
this alteration contributes to the manifestation of ated by adiponectin binding to either of two related cell
obesity-related cardiometabolic diseases53. The mecha­ surface receptors (ADIPOR1 or ADIPOR2)60, which
nisms causing lower adiponectin secretion in obesity is followed by docking of the adaptor protein APPL1
are still under intensive research and probably involve (ref.61) and the stimulation of receptor-intrinsic cerami­
hypoxia, inflammation, endoplasmic reticulum stress dase activity62. The resulting signalling pathway, which
and downregulation of β-adrenergic signalling, which is mediated through the activation of AMP-dependent
is a positive regulator of adiponectin secretion 54–56. kinase, peroxisome proliferator-activated receptor-α
Subcutaneous WAT and BMAT might be especially (PPARα) and other regulatory proteins, leads to meta­
relevant for circulating adiponectin levels. Studies from bolic health-promoting alterations such as decreased
2014 and 2016 showed that adiponectin expression is hepatic gluconeogenesis, increased fatty acid oxidation

512 | SEPTEMBER 2019 | volume 15 www.nature.com/nrendo


Reviews

Table 2 | Regulation, signalling and function of adipose tissue hormones


Endocrine factor Regulation of secretion Molecular signalling Physiological functions
(receptor)
Adipocyte-enriched endocrine factors
Leptin • Increased by fat mass Receptor ligand (LEPR) CNS signalling
• Decreased by β-adrenergic • Increased starvation response
signalling • Decreased appetite
• Decreased by prolonged • Increased energy expenditure
fasting • Decreased heat loss
• Increased hepatic gluconeogenesis
• Decreased insulin resistance
Peripheral signalling
• Decreased insulin secretion
• Decreased atherogenesis
Adiponectin • Increased by β-adrenergic Receptor ligand • Decreased inflammation
signalling (ADIPOR1, ADIPOR2) • Increased insulin sensitivity
• Decreased by endoplasmic • Increased fatty acid catabolism
reticulum stress • Decreased gluconeogenesis
• Decreased by oxidative stress
• Decreased by obesity
Adipsin • Decreased by insulin Complement factor D, • Decreased inflammation by
• Decreased by obesity (mice) generation of clearance of dead cells
complement factor C3a • Increased inflammation by
chemotaxis
• Increased insulin secretion via
C3a receptor
FABP4 Increased by obesity Cytosolic fatty acid • Increased gluconeogenesis
binding, extracellular • Increased insulin resistance
mechanism unknown • Increased atherogenesis
NRG4 • Increased by thermogenic Receptor ligand (ERBB3, • Decreased hepatocyte DNL
activation ERBB4) • Decreased hepatocyte death
• Decreased by obesity • Decreased insulin resistance
• Decreased steatohepatitis
• Increased adipose tissue
angiogenesis
• Increased adipose tissue
innervation
Adipocyte-derived exosomal microRNAs
Exosomal miR-99b Unknown Decreased translation Decreased liver FGF21 expression
Exosomal miR-92a Decreased by thermogenic Decreased translation • Decreased angiogenesis
activation • Decreased obesity
Lipokines
FAHFAs from DNL • Increased by glucose Receptor ligand • Increased glucose tolerance
metabolism (GPR120, GPR40), • Increased GLP1 secretion
• Increased by DNL others? • Increased insulin secretion
Palmitoleate Increased by WAT DNL Unknown • Decreased inflammation
• Decreased atherogenesis
• Increased glucose homeostasis
12,13-diHOME • Increased by cold exposure Unknown • Increased BAT activity
• Increased by exercise • Increased skeletal muscle fatty acid
oxidation
• Increased fatty acid transport
Adipokines also released by hepatocytes
FGF21 Liver Receptor ligand (FGFR1 Liver
• Increased by starvation with β-klotho) • Increased hepatic gluconeogenesis
• Increased by carbohydrate • Increased fatty acid oxidation
overfeeding • Increased ketogenesis
BAT • Increased sympathetic tone
• Increased insulin sensitivity
• Increased by thermogenic
activation BAT
• Increased by transplantation • Improved glucose homeostasis
• Increased by cellular stress • Decreased cardiac hypertrophy
• Increased by hypertension

NATuRe RevIeWS | EndOcRInOlOgy volume 15 | SEPTEMBER 2019 | 513


Reviews

Table 2 (cont.) | Regulation, signalling and function of adipose tissue hormones


Endocrine factor Regulation of secretion Molecular signalling Physiological functions
(receptor)
Adipokines also released by hepatocytes (cont.)
RBP4 Increased by obesity Vitamin A-binding • Increased inflammation
protein, receptor ligand • Increased insulin resistance
(STRA6, TLR4)
IGF1 • Increased by growth hormone Receptor ligand (IGF1R) BAT
• Increased by BAT • Increased glucose homeostasis in
transplantation T1DM
Adipokines also released by immune cells
CXCL14 Increased by thermogenic Receptor ligand • Increased WAT browning
activation (receptor unknown) • Decreased WAT inflammation
IL-6 • Increased by inflammatory Receptor ligand (IL-6RA, • Increased and/or decreased
cytokines glycoprotein 130) inflammation
• Increased by lipolysis • Increased hepatic gluconeogenesis
• Increased by obesity
Resistin Increased by inflammation Receptor ligand (TLR4, • Increased inflammation
others?) • Increased insulin resistance
Ubiquitously expressed adipokines with endocrine potential
GDF15 • Increased by obesity Receptor ligand • Decreased inflammation
• Increased by severe illness (GFRAL) • Increased glucose homeostasis
• Increased by IL-4 and IL-13 • Pleiotropic effects
• Increased by oxidative stress
NAMPT/visfatin • Increased by obesity NAD+-synthesizing • Increased lymphocyte growth
• Increased by insulin resistance enzyme, receptor • Increased inflammation
ligand? • Increased apoptosis
Omentin Decreased by hyperinsulinemia Lectin, receptor ligand? • Decreased insulin resistance
• Decreased atherogenesis
Vaspin Increased by obesity Protease inhibitor Decreased insulin resistance
ADIPOR, adiponectin receptor protein; BAT, brown adipose tissue; CNS, central nervous system; CXCL14, C-X-C motif chemokine
ligand 14; 12,13-diHOME, 12,13-dihydroxy-(9Z)-octadecenoic acid; DNL, de novo lipogenesis; FABP4, fatty acid-binding protein 4;
FAHFAs, fatty acid esters of hydroxy fatty acids; FFA, free fatty acid; FGF21, fibroblast growth factor 21; GDF15, growth differentiation
factor 15; GFRAL, GDNF family receptor-α-like; GLP1, glucagon-like peptide 1; IGF1, insulin-like growth factor 1; LEPR, leptin
receptor; NAMPT, nicotinamide phosphoribosyltransferase; NRG4, neuregulin 4; RBP4, retinol-binding protein 4; T1DM, type 1
diabetes mellitus; TLR4, Toll-like receptor 4; WAT, white adipose tissue.

in liver and skeletal muscle, increased glucose uptake in Taken together, the findings show that adiponectin is
muscle and WAT and decreased WAT inflammation53. an endocrine hormone that is released by adipocytes,
Of note, adiponectin also has a protective effect with proven homeostatic functions, particularly in the
on pancreatic β-cells. Local inflammation and stress context of chronic metabolic stress.
induced by the deposition of lipotoxic lipids (for exam-
ple, ceramides and diacylglycerols) are important mecha­ Other adipokines
nisms causing dysfunction63 and partial β-cell loss Adipsin. The first identified adipokine was adipsin,
during progression from simple insulin resistance to which was originally described as a secreted serine pro-
T2DM. Adiponectin receptors are expressed by β-cells, tease expressed in mature adipocytes but not in undif-
and evidence suggests that adiponectin, in line with its ferentiated precursors72. Plasma adipsin is derived from
general anti-inflammatory role, prevents β-cell apoptosis adipose tissues (Fig. 1), which are the major site of its
and dysregulation of insulin secretion in vitro51. Mouse expression73. Adipsin is also known as complement fac-
experiments using a transgenic model of limited β-cell tor D, which is a rate-limiting component of the alterna-
destruction found that adiponectin was essential to pre- tive complement pathway. In humans, the activity of the
vent hyperglycaemia and loss of pancreatic islet mass, alternative complement pathway correlates with adverse
in part by decreasing levels of lipotoxic ceramides63. outcomes, for example in cardiovascular disease74,
Notably, adiponectin supports the regeneration of β-cells indicating that adipsin has the potential to modulate
in vivo by inducing expression of HNF4A, a master inflammatory events in remote organs. Depending on
transcription factor of β-cells63. the context, the alternative pathway might fuel inflam-
Beyond β-cells, adiponectin exhibits strong mation by the production of chemotactic molecules, or
anti-inflammatory and antifibrotic activities in many might dampen inflammation by opsonization and the
organs by acting on macrophages and fibrogenic removal of dead cells.
cells53,64,65. Accordingly, a protective role of adiponectin Studies using mouse disease models suggest that
could be identified in atherosclerosis66, gestational the net outcome of adipsin action depends on dis-
diabetes67–69 and nonalcoholic fatty liver disease70,71. ease severity; models with more pronounced tissue

514 | SEPTEMBER 2019 | volume 15 www.nature.com/nrendo


Reviews

damage favour a proinflammatory role of adipsin75. endocrine functions and accelerates the development of
However, in mouse models of obesity, circulating lev- obesity-related diseases79–81.
els of adipsin progressively decline over time76. In sup-
port of a protective function of adipsin, a 2014 study Neuregulin 4. Neuregulin 4 (NRG4) is an adipokine
found that adipsin-deficient mice fed a high-fat diet that can counteract chronic inflammation and obesity-
developed glucose intolerance. The phenotype could be associated metabolic changes in the liver83,84. This pro-
explained by defective insulin secretion in the presence tein is a member of the epidermal growth factor family
of unaltered β-cell mass and normal insulin sensitivity76. and is highly expressed in brown and white adipo­cytes
Mechanistically, complement factor C3a, which is gen- (Table 1) . In adipose tissue and especially BAT, the
erated in an adipsin-dependent manner, was found to expression of NRG4 is induced in mice on cold expo-
boost glucose-dependent insulin secretion through C3a sure, whereas expression is decreased in obese mice and
receptor-dependent Ca2+ signalling in β-cells76. humans with obesity83.
In humans, moderately increased adipsin levels NRG4 signalling through the epidermal growth
were observed in the plasma of people with obesity factor receptor homologues ERBB3 and ERBB4 in
as compared with control individuals77. Intriguingly, mouse hepatocytes in vivo leads to attenuated DNL83 as
patients with T2DM who required treatment with well as decreased apoptosis and necrosis in the liver84.
recombinant insulin were characterized by lower Consistent with these protective cellular mechanisms,
plasma adipsin levels76. Thus, a decrease in adipsin lev- NRG4-deficient mice fed a diabetogenic high-fat diet
els in the advanced prediabetic state could be involved have a fatty liver phenotype, gain more weight and are
in the progression to overt T2DM in humans. However, more insulin-resistant than controls, whereas transgenic
as it cannot be ruled out that insulin treatment might mice overexpressing NRG4 show the opposite pheno-
have caused lower adipsin levels in this cohort, more type83. Moreover, NRG4-deficient mice display exacer-
human studies are required to confirm that lower bated inflammation and fibrosis when fed a diet designed
activity of endocrine adipsin increases development to induce nonalcoholic steatohepatitis (NASH)84.
of insulin-dependent T2DM. Although the contribution of adipose tissue to NRG4
plasma levels needs to be formally proved, this factor
Fatty acid-binding protein 4. Fatty acid-binding pro- seems to be a hormone that is enriched in thermogenic
tein 4 (FABP4) is a small cytosolic protein that binds adipocytes and that has beneficial effects on hepato-
fatty acids and structurally related lipids and is abun- cytes in the context of obesity-associated cardiometa-
dantly expressed in adipocytes as well as being found bolic disease. However, evidence indicates that NRG4
in macrophages and capillary endothelial cells (Table 1). has strong paracrine effects on endothelial cells and is
Genetic deficiency or pharmacological inhibition of essential for adequate adipose tissue angiogenesis85,86.
FABP4 decreases insulin resistance in obese mice and Moreover, NRG4 might also be relevant for appropri-
also protects mice from atherosclerosis78. Changes in ate BAT innervation87. In addition, although NRG4 is
intracellular lipid metabolism, in particular decreased expressed at a much lower level in the liver than in adi-
lipolysis and increased de novo lipogenesis (DNL), have pose tissue83, potential exists for hepatic NRG4 to act
been identified as potential mechanisms protecting in a paracrine fashion. Thus, adipose tissue-derived
FABP4-deficient mice from insulin resistance (described NRG4 acting on the liver is probably not the only mech-
later); however, strong evidence also exists for a role of anism for how this adipokine confers systemic insulin
extracellular FABP4 in cardiometabolic disease78. sensitivity. Anti-inflammatory effects and a healthy
FABP4 is released from adipocytes in response to the adipo­kine profile established through local action88 are
induction of lipolysis through non-classical secretory probably also relevant for the systemic effects of NRG4.
mechanisms78,79. Obese mice exhibit increased circulat- Tissue-selective knockdown of NRG4 or its receptors
ing levels of FABP4, and plasma FABP4 concentration will certainly shed light on the biology of this exciting
is increased in conditions of obesity, insulin resis­ novel adipokine, especially to differentiate between its
tance, T2DM and cardiovascular risk in humans78,80. paracrine and endocrine actions.
Infusion of recombinant FABP4 increases hepatic
gluconeogenesis in FABP4-deficient mice, whereas Lipokines
antibody-mediated neutralization of FABP4 in obese Adipose tissues are quantitatively important sites of
mice decreases gluconeogenesis and improves insu- fatty acid metabolism. In interplay with other organs, fatty
lin resistance80,81. Furthermore, in vitro, recombinant acids are relevant not only as energy sources or bio-
FABP4 triggers proatherogenic responses in vascular synthetic precursors but also as regulatory factors. For
smooth muscle and endothelial cells82. example, adipose tissue-derived FFAs induce the expres-
The mechanism of action of FABP4 is unclear; how- sion of catabolic enzymes in hepatocytes by binding
ever, it might involve direct effects through interaction and activating PPARα, thereby linking WAT lipolysis
of the protein with a cell surface receptor, or indirect to metabolism in the fasting state liver89. Beyond this
effects via FABP4-dependent delivery of lipokines80. and other general fatty acid-related mechanisms, certain
Furthermore, although circulating FABP4 is likely to fatty acid species and derivatives produced by adipocytes
originate from adipocytes, potentially other cells (in have been found to exert defined physiological effects.
particular macrophages or capillary endothelial cells) Several studies (described later) suggest that these lipid
can contribute to plasma FABP4. Nevertheless, sev- hormones, referred to in this Review as ‘lipokines’, act in
eral independent studies indicate that FABP4 exerts an endocrine fashion (Box 1).

NATuRe RevIeWS | EndOcRInOlOgy volume 15 | SEPTEMBER 2019 | 515


Reviews

Palmitoleate. The first lipid that was postulated to act vehicle and the degree of insulin resistance of the mice
as an endocrine adipose lipokine was palmitoleate used in the experiments102,103.
(16:1Δ9), an abundant monounsaturated fatty acid that Another unresolved issue is the effective concentra-
is mostly derived from DNL90 (Fig. 1). Typically, the pal- tion of FAHFAs needed to activate GPR120 and GPR40
mitoleate content of WAT and levels of palmitoleate in vitro, which is considerably higher than the concen-
in the plasma FFA pool are decreased in obese and tration present in mouse plasma97,98,101. Thus, the role of
insulin-resistant mice, owing to decreased adipose tissue FAHFAs as endocrine, antidiabetic regulators remains
DNL8. As palmitoleate exhibits anti-inflammatory and controversial, and more research is needed, especially
insulin-sensitizing effects in vitro91,92 and genetic inter- to identify and corroborate molecular FAHFA targets.
ventions that increase WAT DNL in obese mice increase
systemic insulin sensitivity90,93, it has been proposed 12,13-Dihydroxy-(9Z)-octadecenoic acid. 12,13-
that palmitoleate is a lipokine that increases metabolic Dihydroxy-(9Z)-octadecenoic acid (12,13-diHOME)
health90. Supporting this concept, dietary palmitoleate was identified through an unbiased lipidomic approach
supplementation reduces atherosclerosis92, improves glu- as a lipokine the level of which is selectively increased
cose homeostasis90,94, and suppresses liver inflammation95 in the plasma of humans and mice that are exposed to
in mouse disease models. cold104 (Fig. 1). Notably, BAT was identified as the source
Whether a decrease in the palmitoleate content of circulating 12,13-diHOME in mice. This conclu-
of WAT is an important step in the development of sion is based on the observation that the expression
obesity-associated insulin resistance in humans is an of 12,13-diHOME-synthesizing enzyme, bifunctional
open question. In people with obesity and insulin resist- soluble epoxide hydrolase 2, is specifically induced
ance, WAT palmitoleate content is not substantially in BAT depots on cold exposure104. In both mice and
altered as compared with healthy control individuals. humans, plasma 12,13-diHOME concentration corre-
This observation can be explained by the finding that, lated with the volume of active BAT104. Functionally,
unlike in mice, the enzyme catalysing the final step in 12,13-diHOME boosted energy expenditure and stimu­
palmitoleate synthesis, Δ9-desaturase, is not downregu- lated fatty acid uptake in BAT, but not in other organs,
lated in WAT of obese humans96. Thus, there is no sys- probably through the fatty acid transporters CD36
temic palmitoleate deficiency in individuals with obesity and FATP1 (ref.104). Together, these data characterize
and insulin resistance and, consequently, no major con- 12,13-diHOME as a BAT-derived lipokine that augments
tribution to the development of cardiometabolic diseases. adaptive thermogenesis in a paracrine fashion.
Nevertheless, palmitoleate is probably anti-inflammatory A 2018 publication found that the concentration of
in humans, and supplementation might have beneficial 12,13-diHOME is elevated in the plasma of humans and
effects in chronic inflammatory diseases. mice under conditions of moderate-intensity exercise and
that 12,13-diHOME stimulates fatty acid uptake and oxi-
Fatty acid esters of hydroxy fatty acids. Fatty acid esters dation in skeletal muscle105. The study authors showed
of hydroxy fatty acids (FAHFAs) are a novel class of that 12,13-diHOME is primarily produced by BAT; how-
lipids that are linked to adipose tissue DNL97 (Fig. 1). ever, the factor also acts on muscle, thereby providing
Genetic mouse models with suppressed DNL in multiple evidence for an endocrine action. Taken together, the
organs or selectively in adipocytes have decreased levels findings show that 12,13-diHOME is a novel lipokine
of FAHFA species (containing fatty acids derived from that seems to be important for providing fatty acids to
DNL) in adipose tissues and plasma97,98. In both mice organs under conditions of high energy demand, through
and humans, WAT and plasma levels of such FAHFAs cellular mechanisms that need to be further explored.
correlate with systemic and adipocyte insulin sensitiv-
ity97,99. These results led to the model that the systemic Adipocyte-derived exosomal microRNAs
decrease of the levels of WAT DNL-derived FAHFAs in MicroRNAs (miRNAs) are regulatory non-coding
obesity contributes to the development of general insulin RNAs that are shared between cells primarily through
resistance and the deterioration of metabolism. exosomes 106. Evidence is accumulating for a role of
Mechanistic studies performed to provide evidence exosomal miRNAs secreted from WAT and BAT in
for the aforementioned model focused on positional metabolic regulation107–109 (Fig. 1). A major proportion
isomers of palmitic acid hydroxystearic acid (PAHSA), of plasma miRNAs originate from adipose tissues, as
which are an abundant subgroup of WAT FAHFAs97. For indicated by a decrease in the levels of many miRNA
example, oral gavage of 5-PAHSA or 9-PAHSA improves species in the plasma of mice with adipocytes deficient
glucose tolerance in insulin-resistant mice97,98. At the cel- in the miRNA-generating enzyme Dicer, which is a
lular level, activation of the fatty acid receptor GPR120 phenotype that can be rescued by adipose tissue trans-
increases adipocyte glucose transport. Moreover, GPR40 plantation108. Of note, BAT transplantation reversed
activation and the secretion of both glucagon-like pep- the plasma increase of the endocrine hormone fibro-
tide 1 and insulin from intestinal L cells and pancreatic blast growth factor 21 (FGF21) that was observed in
Exosomes β-cells, respectively, were identified as potential mecha­ the Dicer-deficient mice and this effect was linked to
Small cytosol-containing nisms mediating the beneficial metabolic effects of exosomal miR-99b downregulating liver Fgf21 mRNA.
membrane vesicles that are PAHSAs97,100. The effects of PAHSAs administered by In patients with lipodystrophy, a greater number of
released by one cell to be
taken up by other cells and
oral gavage in mice on oral glucose tolerance were, how- miRNAs in the circulation were found to be downreg-
release their content, for ever, not observed by another group101. The discrepancy ulated than upregulated, suggesting that adipose tis-
example microRNA. might be due to differences in the lipid content of the sue is also an important source of plasma miRNAs in

516 | SEPTEMBER 2019 | volume 15 www.nature.com/nrendo


Reviews

humans108. In healthy volunteers exosomal miR-92a is endothelial cells, preadipocytes, macrophages and var-
released by brown adipocytes and plasma levels inversely ious immune cell types that are present throughout the
correlate with human BAT activity measured by meta­ body. Here we describe factors for which an endocrine
bolic imaging107, indicating the potential of plasma function has been demonstrated and that are expressed
miRNAs as a biomarker for BAT activity. In summary, to a certain degree also outside adipose tissue. Cytokines
evidence suggests that WAT and BAT are an important and chemokines are not discussed, as they probably
source of exosomal miRNAs; however, more studies are exert most or all of their functions as paracrine medi-
needed to understand the cell biology and regulation of ators. Exceptions are interleukin-6 (IL-6) and C-X-C
the secretion process, as well as to define the specific motif chemokine ligand 14 (CXCL14), for which endo-
endocrine function of miRNAs in metabolism. crine roles are supported by experimental data. As the
To conclude this section, there are a number of contribution of adipose tissue to the effects of the factors
endocrine factors that are released by white and brown discussed is not self-evident, special focus will be placed
adipocytes (Fig. 1). These hormones regulate systemic on data that suggest a specific contribution of adipose
energy metabolism and influence the development of tissues versus other organs as the source of the respective
cardiometabolic diseases by modulating organ-specific endocrine factor.
functions (Figs 1,3).
Adipokines also released by hepatocytes
Factors also secreted by other organs Fibroblast growth factor 21. The endocrine hormone
Most adipose tissue-derived regulatory secreted factors FGF21 has attracted a lot of attention owing to its ben-
are not exclusively produced by adipocytes but are also eficial pharmacological action on metabolism in obese
produced in other organs or by cells of the adipose tissue rodents and primates. In mice, both adipose tissue and
stroma–vascular fraction (Table 1). The latter includes brain are important target organs that mediate major
metabolic effects (weight loss and decreased plasma
glucose and triglyceride levels) of FGF21 administered
β-Cell dysfunction and/or diabetes mellitus Protective in therapeutic doses110–112. The liver is the primary or
Adiponectin, adipsin only source of plasma FGF21 under basal conditions
Detrimental in mice as well as in humans113,114. The production of
β-Cell eNAMPT hepatic FGF21 is induced on prolonged fasting or pro-
tein restriction, and in this context it increases hepatic
Obesity, insulin resistance and/or Protective gluconeogenesis, fatty acid oxidation and ketogenesis
diabetes diabetes mellitus Leptin, adiponectin, NRG4, palmitoleate, via stimulation of the hypothalamus–pituitary–­
FAHFAs, 12,13-diHOME, FGF21, IGF1, adrenal axis and sympathetic tone110,115. Paradoxically,
Insulin CXCL14, GDF15, vaspin
receptor the production of hepatic FGF21 is also induced by
Detrimental carbo­hydrate overfeeding, and under this condition
FABP4, RBP4, resistin, eNAMPT
it supports the disposal of dietary glucose via acute
Context dependent
Adipocyte IL-6 insulin-sensitizing effects mediated through FGF21
receptors in adipose tissue110,116. Thus, FGF21 produced
in the liver fine-tunes metabolism during both fasting
Nonalcoholic fatty liver disease Protective and acute overfeeding by acting on brain and adipose
Adiponectin, NRG4, palmitoleate
tissue, respectively.
Detrimental
Liver Resistin
The role of adipose tissue-derived FGF21 is more
Context dependent
complex and controversial. FGF21 produced in adipose
IL-6 tissue might not be important in obesity-associated
insulin resistance, as adipose tissue-specific FGF21-
deficient mice that are fed a high-fat diet did not show
Cardiovascular disease (atherosclerosis, Protective decreased circulating FGF21 levels or insulin resistance,
cardiac hypertrophy and so on) Leptin, adiponectin, palmitoleate, FGF21, in contrast to liver-specific FGF21-deficient mice114. By
omentin, vaspin
Blood vessel contrast, adipose tissue-derived FGF21 seems to be rele-
Detrimental
FABP4, eNAMPT vant under thermogenic stimulation. In mice exposed to
cold, FGF21 expression in BAT and subcutaneous WAT
Fig. 3 | Functional relevance of adipokines and lipokines in cardiometabolic is substantially increased; however, this effect usually
diseases. Various adipokines and lipokines have been found to exert either protective or occurs without any increase in plasma FGF21 levels116–118.
detrimental effects on the development of cardiometabolic diseases. Causal, disease- A 2017 study that used tissue-specific FGF21 knock-
modifying roles of the hormones presented have been demonstrated by genetic or down indicates that expression and autocrine signalling
pharmacological intervention studies in rodents, as described in detail in the text. Of note, of FGF21 in adipose tissue is required for the proper
for most of the factors, only a limited number of proof-of concept studies have been
development of thermogenic beige adipocytes in WAT
performed. The intensively studied hormone IL-6 displayed variable, context-dependent
results, which can be explained by cell type-specific modulation of inflammatory and
of cold-exposed mice119.
metabolic pathways. CXCL14, C-X-C motif chemokine ligand 14; 12,13-diHOME, Of note, other studies provide evidence for endo-
12,13-dihydroxy-(9Z)-octadecenoic acid; eNAMPT, extracellular nicotinamide phosphori- crine roles of BAT-derived FGF21. One study reports
bosyltransferase; FABP4, fatty acid-binding protein 4; FAHFAs, fatty acid esters of hydroxy that the severely dysfunctional BAT of UCP1-deficient
fatty acids; FGF21, fibroblast growth factor 21; GDF15, growth differentiation factor 15; mice showed a strong induction of Fgf21 expression
IGF1, insulin-like growth factor 1; NRG4, neuregulin 4; RBP4, retinol-binding protein 4. and elevated plasma FGF21 levels, as compared with

NATuRe RevIeWS | EndOcRInOlOgy volume 15 | SEPTEMBER 2019 | 517


Reviews

wild-type controls120. In another study, transplantation However, increasing plasma RBP4 levels via hepatic
of BAT improved the glucose homeostasis of acceptor overexpression does not affect insulin sensitivity in
wild-type mice with high-fat diet-induced obesity and mice131, whereas adipose tissue-specific overexpression
insulin resistance. This phenotype was accompanied by does not alter plasma RBP4 levels but induces adipocyte
substantially elevated plasma FGF21 levels as a result of hypertrophy and inflammation in WAT132. Thus, strong
increased FGF21 expression that was observed in both evidence exists for a paracrine action of adipose tissue
transplanted and endogenous BAT. The effect was absent RBP4 in insulin resistance, whereas more research is
when donor BAT was deficient in IL-6, indicating an needed to elucidate the endocrine functions of hepatic
essential role of this cytokine in FGF21 induction121. and potentially adipose tissue-derived RBP4.
Further evidence for an endocrine role of BAT-derived
FGF21 was provided by a mouse hypertension model, Adipokines also released by immune cells
which had increased plasma FGF21 levels. BAT surgi- C-X-C motif chemokine ligand 14. CXCL14 is a chemo­
cal removal and BAT-specific knockdowns provided kine expressed by various immune and epithelial cells
strong evidence that BAT-derived FGF21 acted on the throughout the body, lacks a known receptor133, and
heart to diminish cardiac hypertrophy122. Taken together, was proposed in 2018 as a novel endocrine BAT adipo­
BAT can become a source of circulating FGF21 under kine134. In comparison with controls housed at ambient
severe disease conditions, whereas under milder physio­ temperature, cold-exposed rodents showed increased
logical stress ranging from long-term fasting to obe- expression of CXCL14 in brown and white adipocytes,
sity, adipose tissue FGF21 seems to act primarily in a as well as increased release of CXCL14 from BAT as
paracrine fashion. shown by arterial–venous difference. Conversely, plasma
and adipose tissue CXCL14 levels were decreased in
Insulin-like growth factor 1. Under basal conditions, the a high-fat diet-induced mouse model134. CXCL14-
liver secretes abundant amounts of insulin-like growth deficient mice had compromised adaptive thermogen-
factor 1 (IGF1) in a growth hormone (GH)-dependent esis and impaired glucose tolerance, whereas systemic
manner. Of note, IGF1 production is also induced under CXCL14 administration increased the thermogenic
basal conditions by GH in WAT and BAT123. However, cir- capacity of BAT, increased WAT browning and amelio-
culating IGF1 levels and liver Igf1 expression are almost rated insulin resistance in obese mice. Adipose tissue
undetectable in GH receptor-deficient mice124, despite recruitment of anti-inflammatory and prothermogenic
substantial residual Igf1 expression in BAT and WAT123, M2 macrophages was found as a mechanism conferring
thereby demonstrating liver is the dominant source of CXCL14-dependent WAT browning134. In summary,
circulating IGF1. Notably, considerably increased plasma in rodents, CXCL14 is an adipokine released from
IGF1 levels and improved glucose homeostasis were BAT that can increase thermogenic differentiation in
observed in BAT transplantation experiments from donor WAT through M2 macrophage polarization. Research
wild-type mice to an acceptor mouse model of type 1 addressing the expression patterns in humans, the
diabetes mellitus (T1DM)125. Although other hormonal effects of BAT-derived CXCL14 on other organs and
changes observed in this study could explain the improve- immune cells and metabolic characterization of mice
ment in glucose homeostasis (for example, decreased IL-6 with adipocyte-specific knockdown of CXCL14 will
levels as discussed later), this study showed that trans- help clarify the role of this novel BAT-derived adipokine.
planted BAT can be a major source of plasma IGF1 that
probably acts as an endocrine adipokine125. Interleukin-6. IL-6 is a cytokine that is secreted by WAT
and BAT but also by a number of non-adipose tissues,
Serum retinol-binding protein 4. Hepatocytes and adi- especially the skeletal muscle in the context of exer-
pocytes secrete serum retinol-binding protein 4 (RBP4), cise135,136. The signalling pathways of IL-6 are complex
which is the main plasma carrier for retinol (vitamin A). and involve both a transmembrane form and a soluble
Plasma RBP4 levels are frequently increased in humans form of IL-6 receptor (IL-6RA), which both interact
with obesity and insulin resistance, and they correlate with with the broadly expressed co-receptor glycoprotein
RBP4 expression in human visceral WAT16,30,126. RBP4 130 (ref.136). IL-6 is released from WAT into the systemic
directly induces insulin resistance in both muscle tissue circulation of humans, as determined by the measure-
and adipocytes, which is mediated through the RBP4 ment of arterial–venous differences seen even in healthy
receptor STRA6 (ref.127). This factor also confers systemic individuals137, and the level of this factor is frequently
insulin resistance that is mediated by Toll-like receptor 4 elevated in the blood of individuals with obesity and/or
(TLR4)-dependent signalling in macrophages128. Thus, diabetes mellitus.
compelling evidence exists that RBP4 is a contributing The secretion of IL-6 is induced in WAT by various
factor for insulin resistance in obesity. factors, including proinflammatory cytokines and exces-
One important open question is the extent to which sive adipocyte lipolysis138,139. Adipose tissue-derived IL-6
adipose-derived RBP4 contributes to circulating RBP4. has been proposed to act on the liver. This hypothesis
A 2017 study found that plasma RBP4 levels are unde- is based on the observation that hepatic insulin resist-
tectable in lean and obese hepatocyte-specific RBP4- ance and impaired glucose tolerance in obese mice were
deficient mice129, suggesting that RBP4 is an endocrine accompanied by a strong increase in IL-6 production
hepatokine but not an adipokine. Decreasing plasma in WAT and these detrimental metabolic effects were
RBP4 levels achieved with the synthetic retinoid fen- abolished by interventions selectively preventing this
retinide increases insulin sensitivity in obese mice130. IL-6 induction139,140. However, IL-6 and other cytokines

518 | SEPTEMBER 2019 | volume 15 www.nature.com/nrendo


Reviews

can also originate from Kupffer cells and other immune WAT, GDF15 can prevent excessive inflammation and
cells in the liver. improve glucose metabolism153. Studies suggest that
Mouse studies using cell type-specific knockdown GDF15 mediates metabolic effects by acting on the neuro­
of IL-6RA showed variable and unanticipated results. nal brainstem receptor GDNF family receptor-α-like
For example, hepatocyte-specific IL-6RA-deficient (GFRAL)154–156. However, in obesity, GDF15 mRNA is
mice showed systemic insulin resistance and pro- increased not only in WAT but also in the liver and pos-
nounced hepatic inflammation, even when fed a nor- sibly other organs157. Thus, the contribution of adipose
mal diet, demonstrating the anti-inflammatory effects tissue to circulating GDF15 in this context is not clear,
of IL-6 in the liver141. Similarly, myeloid cell-specific and overall the status of GDF15 as an endocrine adipo­
IL-6RA-deficient obese mice showed a poorer meta- kine is not well established. Nevertheless, recombinant
bolic phenotype than obese controls associated with GDF15 infusion reduces body weight and corrects
WAT macrophages polarized towards a proinflamma- many obesity-related metabolic derangements in rodent
tory M1 phenotype142,143. By contrast, T cell-specific or and primate models of obesity, thereby indicating
natural killer cell-specific IL-6RA deficiency improved therapeutic potential157.
insulin resistance in obese mice144,145. Thus, IL-6 can act
in both a proinflammatory and an anti-inflammatory Nicotinamide phosphoribosyltransferase. Nicotinamide
fashion, depending on cell type and context. Given these phosphoribosyltransferase (NAMPT; also known as
complex functions, it is not surprising that the role of visfatin) is a protein that functions both intracellularly
WAT-derived IL-6 in hepatic glucose metabolism and and extracellularly. The extracellular function was first
insulin resistance is still controversial135,136. identified through its activity as a bone marrow-derived
B cell growth factor158. In 2005, researchers discovered
Resistin. Resistin is a protein that is secreted from mouse that this factor is expressed in human visceral adipose
adipocytes and macrophages, and its level is increased tissue and renamed it ‘visfatin’159. In the same publica-
in the plasma of obese mice as compared with lean con- tion, the protein was reported to have insulin-mimetic
trols146,147. Resistin accelerates insulin resistance as well properties, which could not be confirmed, leading to
as the development of inflammatory metabolic diseases, the retraction of the article. The visfatin story took an
including NASH, by acting on many cell types, includ- unexpected turn when scientists discovered that the
ing adipocytes, hepatocytes, myocytes, macrophages, protein is NAMPT, which is the rate-limiting enzyme
endothelial cells and hypothalamic neurons146. The pat- in the salvage pathway of nicotine amide dinucleotide
tern recognition receptor TLR4 mediates at least some (NAD+) synthesis, thereby identifying an important
of the proinflammatory effects of resistin148. However, no intracellular function of the protein159.
specific receptor has been identified so far, which limits As NAMPT is widely expressed, it remains unclear
functional studies of this adipokine. whether adipose tissue is an important source of circu-
Importantly, human resistin is derived from circu- lating extracellular NAMPT (eNAMPT). Experiments
lating monocytes and tissue-resident macrophages, to address this question using adipocyte-specific
but not adipocytes146,149. Nevertheless, plasma levels of NAMPT-deficient mice were inconclusive owing to
the hormone also increase with obesity in humans146. severe adipose tissue dysfunction in these animals160.
Notably, a humanized mouse model expressing human Nevertheless, plasma NAMPT levels are positively
resistin exclusively in macrophages showed increased associated with obesity and insulin resistance in many
insulin resistance and inflammation as compared with clinical studies159. Moreover, eNAMPT has various
control animals150. Thus, human resistin plays a role in activities, including some that implicate it in the patho-
obesity-associated inflammation and insulin resistance, genesis of chronic diseases such as atherosclerosis161
but in humans the contribution of adipose tissue to plasma and diabetes162.
resistin levels is probably less important than in mice. Importantly, eNAMPT increases the proinflamma-
tory actions of macrophages and neutrophils, boosts the
Ubiquitously expressed adipokines activation and proliferation of lymphocytes and exerts
Growth differentiation factor 15. Growth differentiation detrimental effects on pancreatic β-cells159,162. Of note,
factor 15 (GDF15; also known as macrophage inhibitory although eNAMPT activates well-established signal-
cytokine 1) is a member of the transforming growth fac- ling pathways related to inflammation and apoptosis,
tor superfamily with anti-inflammatory properties. In a signalling receptor has not yet been identified. It has
humans, GDF15 plasma levels are increased in many been speculated that the enzymatic activity of eNAMPT,
pathological conditions, including cancer, cardiovascular which regulates adenine nucleotide signalling, medi-
disease and kidney failure, and GDF15 levels are positively ates the observed cellular effects159. More intervention
associated with the risk of death151. Many organs can pro- studies that separate intracellular functions from extra-
duce GDF15 (for example, liver and skeletal muscle). cellular functions (for example, using inactivating anti­
However, this factor is not expressed in these organs under bodies162) are needed to understand the role of eNAMPT
basal conditions and is induced only on oxidative stress, in metabolic regulation and cardiometabolic disease.
proinflammatory cytokine action or other stimuli151.
GDF15 is also secreted by preadipocytes and adipo- Omentin. Omentin (also known as intelectin 1) is a
cytes152, and serum levels are increased in humans with lectin-like protein that is expressed in the small and large
obesity and in obese mice. As a downstream effector intestine, where it is involved in microbial surveillance163.
of the anti-inflammatory cytokines IL-4 and IL-13, in Furthermore, omentin is produced by endothelial and

NATuRe RevIeWS | EndOcRInOlOgy volume 15 | SEPTEMBER 2019 | 519


Reviews

other stroma–vascular cells in various organs. The tackle obesity-related metabolic dysfunction. Proving
name ‘omentin’ is derived from the high expression of this concept, leptin treatment was found to be effective
this factor in omental (that is, visceral) WAT compared in humans with obesity caused by rare leptin loss-of-
with subcutaneous human WAT164. Notably, mice do not function mutations33. However, one major hurdle in the
express appreciable amounts of omentin in adipose tis- development of leptin as a general antiobesity drug is
sues, which has hampered mechanistic studies address- the presence of leptin resistance in individuals with
ing the physiological role of adipose-derived omentin165. obesity and in obese rodents, where typically very high
Observational studies have established that low omen- plasma levels of the hormone are present. Leptin resist-
tin plasma levels in humans are associated with insulin ance is a complex phenomenon that involves defects in
resistance and a higher risk of cardiovascular disease165. the uptake of leptin into the cerebrospinal compartment,
Moreover, in humans, adipose tissue production of as well as the attenuation of leptin signalling in central
omentin is suppressed by insulin, which explains the neurons175. Notably, potential approaches to overcome
decrease of circulating omentin levels in hyperinsuli- leptin resistance in obesity have been identified, including
naemic insulin-resistant states166. Although no specific treatment with the islet hormone amylin176–178 and agents
cell surface receptor has been identified, omentin elicits that restore leptin signalling by decreasing intracellu-
distinct downstream signalling events, which explain the lar stress in central neurons179. Although the potential
beneficial metabolic effects165 and suggest therapeutic adverse effects of leptin therapy, such as liver fibrosis180,
potential. need to be kept in mind, the leptin signalling pathway
is amenable to pharmacological intervention, and leptin
Vaspin. Vaspin (also known as serpin A12) is an sensitizing may be a viable option for the treatment of
insulin-sensitizing protease inhibitor, the level of which common metabolic diseases.
is frequently but not always increased in the circulation
of individuals with obesity16,167. Vaspin was originally Adiponectin-based therapies
identified as a visceral WAT-specific adipokine in dia- Given its pleiotropic beneficial effect in cardiometa-
betic rats167. Subsequent work showed that in humans bolic diseases (as identified in rodent disease models),
vaspin was detectable only in WAT of individuals with adipo­nectin signalling is an attractive therapeutic target.
obesity but was not confined to visceral WAT168; vaspin The levels of circulating adiponectin can be elevated by
was also enriched in the stroma–vascular fraction of pharmacological intervention using PPARγ agonists.
adipose tissue169. As vaspin is widely expressed, and Moreover, adiponectin mediates at least part of the result-
no adipose tissue-specific deficient models have been ing positive effects of PPARγ agonist treatment on insu-
reported so far, it remains unclear whether circulating lin resistance, development of fatty liver and other organ
vaspin is derived from adipocytes. Nevertheless, animal disturbances, as comparisons of adiponectin-deficient
studies demonstrate promising beneficial roles of vaspin and wild-type mice demonstrated181,182. Adiponectin
in diabetes and cardiovascular disease167. Future work receptors were also targeted directly in rodents using
should focus on the mechanism of action of vaspin, AdipoRon, an orally available small-molecule ADIPOR1
which may involve inhibition of specific proteases and ADIPOR2 dual agonist183. AdipoRon administered
(for example, kallikrein 7)170. to obese mice not only improved insulin resistance183 but
Taken together, the findings show that a large num- also slowed the development of inflammatory disorders
ber of regulatory molecules with divergent activities are such as generalized fibrosis184, providing proof of con-
secreted from adipose tissues. Most of these molecules cept for adiponectin-specific therapies. A general caveat
are also produced by other organs or by other cell types is that even though high adiponectin levels are strongly
in adipose tissues (Table 1), and in many cases the exact associated with insulin sensitivity and a low risk of devel-
contribution of adipocytes needs to be established. oping T2DM in humans, a causal role of adiponectin and
salutary effects of high adiponectin levels on mortality
Therapeutic aspects could not be confirmed in large genetic studies185. These
Leptin-based therapies findings cast doubt on the general translatability of the
As described previously, leptin is a hormone essential rodent data to humans, and more research is warranted
for the regulation of energy metabolism and major to resolve this paradox.
endocrine axes. Accordingly, recombinant leptin can be
used to treat endocrine dysfunctions in hypoleptinae- Other adipokine-based therapies
mic conditions that are present in extremely lean women Although many adipokines and other adipose tissue hor-
with amenorrhoea171 and also in patients with lipodys- mones are associated with cardiometabolic diseases and
trophy172. In addition, leptin administration normalizes are promising as biomarkers186, none have as yet as much
plasma glucose levels in rodent T1DM models that have therapeutic validation as leptin. Therapeutic targeting
low leptin levels173. The mechanisms described remain of molecules secreted from adipose tissue is generally
unclear, but leptin may act by correcting glucagon hyper- difficult, and a potential reason for this is that many
secretion or by suppressing cortisol secretion174, or both. exert pleiotropic, modulatory functions. This fact is not
However, leptin deficiency is less pronounced in human ideal with regard to therapeutic efficacy, especially in the
T1DM, and leptin treatment might not be effective in context of complex cardiometabolic diseases that require
patients with T1DM174. long-term treatment, as both pleiotropic effects and
The weight-lowering and insulin-sensitizing effects sustained dosing increase the risk of adverse effects.
of leptin make it an attractive therapeutic approach to For example, the administration of NRG4 might exert

520 | SEPTEMBER 2019 | volume 15 www.nature.com/nrendo


Reviews

beneficial cardiometabolic effects but at the same Conclusions


time increase the risk of breast cancer development A large number of plasma hormones originate from
through permanent activation of the receptors ERBB3 WAT and BAT, and are secreted depending on the energy
and ERBB4 (ref.187). In addition, the development of and disease states of these adipose tissues (Fig. 2; Table 2).
oral drugs for peptide factors such as adipokines is The relevance of these factors is especially exemplified
challenging, and thus pharmaceutical efforts to develop by the presence of hypertrophic adipocytes in obesity,
endocrine therapies based on these molecules are where adipose tissue-derived proinflammatory mol-
limited so far. ecules can affect β-cell function, obesity-associated
Nevertheless, many adipokines are of great thera- insulin resistance and the development and progres-
peutic interest. Treatment with pharmacological doses sion of diabetes, NASH and various cardiovascular
of FGF21 corrects elevated plasma levels of glucose and diseases (Fig. 3).
triglycerides in rodent models, non-human primates Notably, many adipose tissue-derived hormones are
and patients with diabetes mellitus111,112,188. Moreover, expressed to a substantial degree in other organs, and
a long-acting analogue of FGF21 was found to reduce their endocrine versus paracrine contribution is fre-
liver steatosis in patients with NASH189. GDF15 is quently not defined. This issue could be addressed by
another promising therapeutic adipokine, as this factor more frequently using in vivo technologies that are based
reduces weight in obese mice and monkey models by on arterial–venous or microdialysis sampling, thereby
suppressing appetite, even in a chronic setting, proba- allowing quantification of secretion from adipose tissue
bly through defined areas in the hindbrain157. Several depots in rodents and humans. Moreover, more
rodent studies indicate that omentin has various ben- research using cell type-specific genetic manipulation
eficial effects in cardiovascular diseases. For example, of these hormones and their receptors is needed to dis-
adenoviral or transgenic expression of human omentin sect systemic and pleiotropic effects. Furthermore, the
attenuates cardiac hypertrophy, reduces atherosclerosis combination of global interventions such as constant
and prevents myocardial injury in mouse models190–192. adipo­kine or lipokine infusion, or neutralizing antibodies
Recombinant vaspin protects rats against myocardial combined with organ-specific receptor knockdown
ischaemia and reperfusion injury193, as well as alleviat- would advance our understanding of adipocyte-derived
ing pulmonary hypertension by inhibiting inflammation hormone endocrine functions. The role of specific cell
and remodelling of the vascular wall194. types can also be investigated by novel nucleic acid-
Harmful adipokines and their effect on metabolism based intervention methods. For example, adenovirus-
and cardiovascular disease can be neutralized by anti- associated virus vectors and small interfering RNAs
bodies, as shown, for example, in the case of FABP4. encapsulated by a glucan shell can be used to target
Long-term treatment of obese mice targeting extracel- adipocytes and WAT-resident macrophages, respec-
lular FABP4 resulted in improved glucose homeostasis tively195,196. Finally, it will be important to translate the
and increased insulin sensitivity, and decreased fat mass proof of concept generated in rodent models to the clini­
as well as liver steatosis81. In summary, administration cal setting and demonstrate feasibility as well as effi-
of therapeutic adipokines and adipokine-neutralizing ciency of therapies based on endocrine signalling of
antibodies has provided encouraging results in rodent adipose tissue hormones in humans.
models; however, for most of them efficacy and safety
in humans needs to be proven. Published online 11 July 2019

1. Zhang, Y. et al. Positional cloning of the mouse obese 9. Bordicchia, M. et al. Cardiac natriuretic peptides act 18. Cannon, B. & Nedergaard, J. Brown adipose tissue:
gene and its human homologue. Nature 372, via p38 MAPK to induce the brown fat thermogenic function and physiological significance. Physiol. Rev.
425–432 (1994). program in mouse and human adipocytes. J. Clin. 84, 277–359 (2004).
2. Karastergiou, K. & Mohamed-Ali, V. The autocrine and Invest. 122, 1022–1036 (2012). 19. Li, Y. et al. Secretin-activated brown fat mediates
paracrine roles of adipokines. Mol. Cell Endocrinol. 10. Crewe, C., An, Y. A. & Scherer, P. E. The ominous triad prandial thermogenesis to induce satiation. Cell 175,
318, 69–78 (2010). of adipose tissue dysfunction: inflammation, fibrosis, 1561–1574 (2018).
3. Lehr, S., Hartwig, S. & Sell, H. Adipokines: a treasure and impaired angiogenesis. J. Clin. Invest. 127, 20. Heine, M. et al. Lipolysis triggers a systemic insulin
trove for the discovery of biomarkers for metabolic 74–82 (2017). response essential for efficient energy replenishment
disorders. Proteomics Clin. Appl. 6, 91–101 11. Saltiel, A. R. & Olefsky, J. M. Inflammatory of activated brown adipose tissue in mice. Cell Metab.
(2012). mechanisms linking obesity and metabolic disease. 28, 644–655 (2018).
4. Ali Khan, A. et al. Comparative secretome analyses of J. Clin. Invest. 127, 1–4 (2017). 21. Bartelt, A. & Heeren, J. Adipose tissue browning and
primary murine white and brown adipocytes reveal 12. Hotamisligil, G. S. Inflammation, metaflammation and metabolic health. Nat. Rev. Endocrinol. 10, 24–36
novel adipokines. Mol. Cell Proteomics 17, immunometabolic disorders. Nature 542, 177–185 (2014).
2358–2370 (2018). (2017). 22. Villarroya, F., Cereijo, R., Villarroya, J.,
5. Seldin, M. M. et al. A strategy for discovery of 13. Boucher, J., Kleinridders, A. & Kahn, C. R. Insulin Gavalda-Navarro, A. & Giralt, M. Toward an
endocrine interactions with application to whole-body receptor signaling in normal and insulin-resistant understanding of how immune cells control brown and
metabolism. Cell Metab. 27, 1138–1155 (2018). states. Cold Spring Harb. Perspect. Biol. 6, a009191 beige adipobiology. Cell Metab. 27, 954–961 (2018).
This study identified lipocalin 5 as a novel (2014). 23. Takeshita, S., Fumoto, T., Naoe, Y. & Ikeda, K.
adipokine regulating muscle mitochondria by using 14. Xu, X. et al. Obesity activates a program of Age-related marrow adipogenesis is linked to
an unbiased computational approach based on lysosomal-dependent lipid metabolism in adipose increased expression of RANKL. J. Biol. Chem. 289,
multiorgan transcriptomics of various mouse tissue macrophages independently of classic 16699–16710 (2014).
strains. activation. Cell Metab. 18, 816–830 (2013). 24. Naveiras, O. et al. Bone-marrow adipocytes as
6. Giordano, A., Smorlesi, A., Frontini, A., Barbatelli, G. 15. Petersen, M. C. & Shulman, G. I. Mechanisms of negative regulators of the haematopoietic
& Cinti, S. White, brown and pink adipocytes: the insulin action and insulin resistance. Physiol. Rev. 98, microenvironment. Nature 460, 259–263 (2009).
extraordinary plasticity of the adipose organ. Eur. J. 2133–2223 (2018). 25. Cawthorn, W. P. et al. Bone marrow adipose tissue is
Endocrinol. 170, R159–R171 (2014). 16. Kloting, N. et al. Insulin-sensitive obesity. Am. J. an endocrine organ that contributes to increased
7. Tchernof, A. & Despres, J. P. Pathophysiology of Physiol. Endocrinol. Metab. 299, E506–E515 circulating adiponectin during caloric restriction.
human visceral obesity: an update. Physiol. Rev. 93, (2010). Cell Metab. 20, 368–375 (2014).
359–404 (2013). 17. Karpe, F. & Pinnick, K. E. Biology of upper-body and 26. Costa, R. M., Neves, K. B., Tostes, R. C. & Lobato, N. S.
8. Scheja, L. & Heeren, J. Metabolic interplay lower-body adipose tissue—link to whole-body Perivascular adipose tissue as a relevant fat depot
between white, beige, brown adipocytes and the phenotypes. Nat. Rev. Endocrinol. 11, 90–100 for cardiovascular risk in obesity. Front. Physiol. 9,
liver. J. Hepatol. 64, 1176–1186 (2016). (2015). 253 (2018).

NATuRe RevIeWS | EndOcRInOlOgy volume 15 | SEPTEMBER 2019 | 521


Reviews

27. Xiong, W. et al. Brown adipocyte-specific PPARγ 51. Dunmore, S. J. & Brown, J. E. The role of adipokines in 76. Lo, J. C. et al. Adipsin is an adipokine that improves
(peroxisome proliferator-activated receptor gamma) beta-cell failure of type 2 diabetes. J. Endocrinol. 216, beta cell function in diabetes. Cell 158, 41–53
deletion impairs perivascular adipose tissue T37–45 (2013). (2014).
development and enhances atherosclerosis in mice. 52. Soedling, H. et al. Limited impact on glucose 77. Maslowska, M. et al. Plasma acylation stimulating
Arterioscler Thromb. Vasc. Biol. 38, 1738–1747 homeostasis of leptin receptor deletion from insulin- protein, adipsin and lipids in non-obese and obese
(2018). or proglucagon-expressing cells. Mol. Metab. 4, populations. Eur. J. Clin. Invest. 29, 679–686 (1999).
28. Antonopoulos, A. S. et al. Detecting human coronary 619–630 (2015). 78. Hotamisligil, G. S. & Bernlohr, D. A. Metabolic
inflammation by imaging perivascular fat. Sci. Transl 53. Fang, H. & Judd, R. L. Adiponectin regulation and functions of FABPs—mechanisms and therapeutic
Med. 9, eaal2658 (2017). function. Compr. Physiol. 8, 1031–1063 (2018). implications. Nat. Rev. Endocrinol. 11, 592–605
29. Guglielmi, V. & Sbraccia, P. Epicardial adipose tissue: 54. Komai, A. M. et al. White adipocyte adiponectin (2015).
at the heart of the obesity complications. Acta exocytosis is stimulated via β3-adrenergic signaling 79. Villeneuve, J. et al. Unconventional secretion of FABP4
Diabetol. 54, 805–812 (2017). and activation of Epac1: catecholamine resistance in by endosomes and secretory lysosomes. J. Cell Biol.
30. Bluher, M. & Mantzoros, C. S. From leptin to other obesity and type 2 diabetes. Diabetes 65, 217, 649–665 (2018).
adipokines in health and disease: facts and 3301–3313 (2016). 80. Cao, H. et al. Adipocyte lipid chaperone AP2 is
expectations at the beginning of the 21st century. 55. Kikai, M. et al. Adrenergic receptor-mediated a secreted adipokine regulating hepatic glucose
Metabolism 64, 131–145 (2015). activation of FGF-21-adiponectin axis exerts production. Cell Metab. 17, 768–778 (2013).
31. Friedman, J. The long road to leptin. J. Clin. Invest. atheroprotective effects in brown adipose 81. Burak, M. F. et al. Development of a therapeutic
126, 4727–4734 (2016). tissue-transplanted apoE−/− mice. Biochem. Biophys. monoclonal antibody that targets secreted fatty
32. Fischer, A. W., Cannon, B. & Nedergaard, J. Res. Commun. 497, 1097–1103 (2018). acid-binding protein aP2 to treat type 2 diabetes.
Leptin-deficient mice are not hypothermic, they are 56. Trayhurn, P. Hypoxia and adipose tissue function Sci. Transl Med. 7, 319ra205 (2015).
anapyrexic. Mol. Metab. 6, 173 (2017). and dysfunction in obesity. Physiol. Rev. 93, 1–21 82. Girona, J. et al. FABP4 induces vascular smooth
33. Farooqi, I. S. & O’Rahilly, S. 20 years of leptin: (2013). muscle cell proliferation and migration through a
human disorders of leptin action. J. Endocrinol. 223, 57. Sulston, R. J. et al. Increased circulating adiponectin in MAPK-dependent pathway. PLOS ONE 8, e81914
T63–70 (2014). response to thiazolidinediones: investigating the role (2013).
34. Boden, G., Chen, X., Mozzoli, M. & Ryan, I. Effect of bone marrow adipose tissue. Front. Endocrinol. 7, 83. Wang, G. X. et al. The brown fat-enriched secreted
of fasting on serum leptin in normal human subjects. 128 (2016). factor Nrg4 preserves metabolic homeostasis through
J. Clin. Endocrinol. Metab. 81, 3419–3423 (1996). 58. Maeda, N. et al. Diet-induced insulin resistance in attenuation of hepatic lipogenesis. Nat. Med. 20,
35. Sinha, M. K. et al. Nocturnal rise of leptin in lean, mice lacking adiponectin/ACRP30. Nat. Med. 8, 1436–1443 (2014).
obese, and non-insulin-dependent diabetes mellitus 731–737 (2002). This study discovered that cold exposure triggers
subjects. J. Clin. Invest. 97, 1344–1347 (1996). 59. Kim, J. Y. et al. Obesity-associated improvements in the induction of NRG4 expression in BAT of mice
36. Francisco, V. et al. Obesity, fat mass and immune metabolic profile through expansion of adipose tissue. and in parallel reduces hepatic DNL through NRG4
system: role for leptin. Front. Physiol. 9, 640 (2018). J. Clin. Invest. 117, 2621–2637 (2007). receptor (ERBB3/ERBB4) signalling, providing
37. Hube, F. et al. Difference in leptin mRNA levels 60. Yamauchi, T. et al. Cloning of adiponectin receptors evidence for a novel endocrine BAT–liver axis.
between omental and subcutaneous abdominal that mediate antidiabetic metabolic effects. Nature 84. Guo, L. et al. Hepatic neuregulin 4 signaling defines an
adipose tissue from obese humans. Horm. Metab. Res. 423, 762–769 (2003). endocrine checkpoint for steatosis-to-NASH
28, 690–693 (1996). 61. Mao, X. et al. APPL1 binds to adiponectin receptors progression. J. Clin. Invest. 127, 4449–4461 (2017).
38. Wrann, C. D. et al. FOSL2 promotes leptin gene and mediates adiponectin signalling and function. 85. Nugroho, D. B., Ikeda, K., Kajimoto, K., Hirata, K. I. &
expression in human and mouse adipocytes. Nat. Cell Biol. 8, 516–523 (2006). Emoto, N. Activation of neuregulin-4 in adipocytes
J. Clin. Invest. 122, 1010–1021 (2012). 62. Vasiliauskaite-Brooks, I. et al. Structural insights into improves metabolic health by enhancing adipose
39. Caron, A., Lee, S., Elmquist, J. K. & Gautron, L. Leptin adiponectin receptors suggest ceramidase activity. tissue angiogenesis. Biochem. Biophys. Res. Commun.
and brain-adipose crosstalks. Nat. Rev. Neurosci. 19, Nature 544, 120–123 (2017). 504, 427–433 (2018).
153–165 (2018). 63. Ye, R., Wang, M., Wang, Q. A. & Scherer, P. E. 86. Nugroho, D. B. et al. Neuregulin-4 is an angiogenic
40. Mantzoros, C. S. et al. Activation of β3 adrenergic Adiponectin-mediated antilipotoxic effects in factor that is critically involved in the maintenance of
receptors suppresses leptin expression and mediates regenerating pancreatic islets. Endocrinology 56, adipose tissue vasculature. Biochem. Biophys. Res.
a leptin-independent inhibition of food intake in mice. 2019–2028 (2015). Commun. 503, 378–384 (2018).
Diabetes 45, 909–914 (1996). 64. Mandal, P., Pratt, B. T., Barnes, M., McMullen, M. R. 87. Christian, M. Transcriptional fingerprinting of
41. Trayhurn, P., Duncan, J. S., Rayner, D. V. & Hardie, L. J. & Nagy, L. E. Molecular mechanism for “browning” white fat identifies NRG4 as a novel
Rapid inhibition of ob gene expression and circulating adiponectin-dependent M2 macrophage polarization: adipokine. Adipocyte 4, 50–54 (2015).
leptin levels in lean mice by the beta 3-adrenoceptor link between the metabolic and innate immune 88. Chen, Z. et al. Nrg4 promotes fuel oxidation and a
agonists BRL 35135A and ZD2079. Biochem. activity of full-length adiponectin. J. Biol. Chem. 286, healthy adipokine profile to ameliorate diet-induced
Biophys. Res. Commun. 228, 605–610 (1996). 13460–13469 (2011). metabolic disorders. Mol. Metab. 6, 863–872
42. Cohen, P. et al. Selective deletion of leptin receptor in 65. Caligiuri, A. et al. Adenosine monophosphate-activated (2017).
neurons leads to obesity. J. Clin. Invest. 108, protein kinase modulates the activated phenotype 89. Montagner, A. et al. Liver PPARalpha is crucial for
1113–1121 (2001). of hepatic stellate cells. Hepatology 47, 668–676 whole-body fatty acid homeostasis and is protective
43. Zeng, W. et al. Sympathetic neuro-adipose connections (2008). against NAFLD. Gut 65, 1202–1214 (2016).
mediate leptin-driven lipolysis. Cell 163, 84–94 66. Okamoto, Y. et al. Adiponectin reduces atherosclerosis 90. Cao, H. et al. Identification of a lipokine, a lipid
(2015). in apolipoprotein E-deficient mice. Circulation 106, hormone linking adipose tissue to systemic
44. Hayes, M. R. et al. Endogenous leptin signaling in the 2767–2770 (2002). metabolism. Cell 134, 933–944 (2008).
caudal nucleus tractus solitarius and area postrema is 67. Qiao, L. et al. Adiponectin deficiency impairs maternal 91. Chan, K. L. et al. Palmitoleate reverses high
required for energy balance regulation. Cell Metab. metabolic adaptation to pregnancy in mice. Diabetes fat-induced proinflammatory macrophage polarization
11, 77–83 (2010). 66, 1126–1135 (2017). via AMP-activated protein kinase (AMPK). J. Biol.
45. Scott, M. M., Williams, K. W., Rossi, J., Lee, C. E. & 68. Cheng, L. et al. Adiponectin deficiency leads to female Chem. 290, 16979–16988 (2015).
Elmquist, J. K. Leptin receptor expression in hindbrain subfertility and ovarian dysfunctions in mice. 92. Cimen, I. et al. Prevention of atherosclerosis by
Glp-1 neurons regulates food intake and energy Endocrinology 157, 4875–4887 (2016). bioactive palmitoleate through suppression of
balance in mice. J. Clin. Invest. 121, 2413–2421 69. Aye, I. L., Rosario, F. J., Powell, T. L. & Jansson, T. organelle stress and inflammasome activation.
(2011). Adiponectin supplementation in pregnant mice Sci. Transl Med. 8, 358ra126 (2016).
46. Denroche, H. C. et al. Disrupted leptin signaling in the prevents the adverse effects of maternal obesity on The study showed that oral supplementation with
lateral hypothalamus and ventral premammillary placental function and fetal growth. Proc. Natl Acad. the lipokine palmitoleate reduces the size of
nucleus alters insulin and glucagon secretion and Sci. USA 112, 12858–12863 (2015). atherosclerotic lesions in apolipoprotein E-deficient
protects against diet-induced obesity. Endocrinology 70. Hu, X. et al. MitoNEET deficiency alleviates mice by relieving cholesterol-induced endoplasmic
157, 2671–2685 (2016). experimental alcoholic steatohepatitis in mice by reticulum stress and inflammatory mechanisms in
47. Xu, J. et al. Genetic identification of leptin neural stimulating endocrine adiponectin-Fgf15 axis. J. Biol. macrophages.
circuits in energy and glucose homeostases. Nature Chem. 291, 22482–22495 (2016). 93. Herman, M. A. et al. A novel ChREBP isoform in
556, 505–509 (2018). 71. Wang, J. et al. Myeloid cell-specific lipin-1 deficiency adipose tissue regulates systemic glucose metabolism.
48. Berglund, E. D. et al. Direct leptin action on POMC stimulates endocrine adiponectin-FGF15 axis and Nature 484, 333–338 (2012).
neurons regulates glucose homeostasis and hepatic ameliorates ethanol-induced liver injury in mice. 94. Yang, Z. H., Miyahara, H. & Hatanaka, A. Chronic
insulin sensitivity in mice. J. Clin. Invest. 122, Sci. Rep. 6, 34117 (2016). administration of palmitoleic acid reduces insulin
1000–1009 (2012). 72. Cook, K. S. et al. Adipsin: a circulating serine protease resistance and hepatic lipid accumulation in KK-Ay
49. Hubert, A. et al. Selective deletion of leptin signaling homolog secreted by adipose tissue and sciatic nerve. Mice with genetic type 2 diabetes. Lipids Health Dis.
in endothelial cells enhances neointima formation and Science 237, 402–405 (1987). 10, 120 (2011).
phenocopies the vascular effects of diet-induced 73. Wu, X. et al. Contribution of adipose-derived factor 95. Guo, X. et al. Palmitoleate induces hepatic steatosis
obesity in mice. Arterioscler. Thromb. Vasc. Biol. 37, D/adipsin to complement alternative pathway but suppresses liver inflammatory response in mice.
1683–1697 (2017). activation: lessons from lipodystrophy. J. Immunol. PLOS ONE 7, e39286 (2012).
Using endothelial cell-specific Lepr-knockout mice, 200, 2786–2797 (2018). 96. Eissing, L. et al. De novo lipogenesis in human fat and
this study identified leptin signalling in endothelial 74. Hertle, E. et al. The alternative complement pathway is liver is linked to ChREBP-beta and metabolic health.
cells as an important mechanism counteracting longitudinally associated with adverse cardiovascular Nat. Commun. 4, 1528 (2013).
obesity-associated neointima formation. outcomes. The CODAM study. Thromb. Haemost. 115, 97. Yore, M. M. et al. Discovery of a class of endogenous
50. Wu, Y., Fortin, D. A., Cochrane, V. A., Chen, P. C. 446–457 (2016). mammalian lipids with anti-diabetic and
& Shyng, S. L. NMDA receptors mediate leptin 75. McCullough, R. L. et al. Complement factor D protects anti-inflammatory effects. Cell 159, 318–332 (2014).
signaling and regulate potassium channel trafficking mice from ethanol-induced inflammation and liver This study describes the discovery of FAHFAs as a
in pancreatic beta-cells. J. Biol. Chem. 292, injury. Am. J. Physiol. Gastrointest. Liver Physiol. 315, novel class of lipokines that are tightly linked to
15512–15524 (2017). G66–G79 (2018). WAT DNL and systemic insulin sensitivity.

522 | SEPTEMBER 2019 | volume 15 www.nature.com/nrendo


Reviews

98. Vijayakumar, A. et al. Absence of carbohydrate 123. Hjortebjerg, R. et al. Insulin, IGF-1, and GH receptors inflammation and insulin resistance. Nat. Commun. 8,
response element binding protein in adipocytes are altered in an adipose tissue depot-specific manner 14803 (2017).
causes systemic insulin resistance and impairs glucose in male mice with modified GH action. Endocrinology 146. Schwartz, D. R. & Lazar, M. A. Human resistin: found
transport. Cell Rep. 21, 1021–1035 (2017). 158, 1406–1418 (2017). in translation from mouse to man. Trends Endocrinol.
99. Hammarstedt, A. et al. Adipose tissue dysfunction is 124. Masternak, M. M. et al. Effects of caloric restriction Metab. 22, 259–265 (2011).
associated with low levels of the novel palmitic acid on insulin pathway gene expression in the skeletal 147. Tan, Y. et al. Antiresistin RNA oligonucleotide
hydroxystearic acids. Sci. Rep. 8, 15757 (2018). muscle and liver of normal and long-lived GHR-KO ameliorates diet-induced nonalcoholic fatty liver
100. Syed, I. et al. Palmitic acid hydroxystearic acids mice. Exp. Gerontol. 40, 679–684 (2005). disease in mice through attenuating proinflammatory
activate GPR40, which is involved in their beneficial 125. Gunawardana, S. C. & Piston, D. W. Insulin-independent cytokines. Biomed. Res. Int. 2015, 414860 (2015).
effects on glucose homeostasis. Cell Metab. 27, reversal of type 1 diabetes in nonobese diabetic mice 148. Benomar, Y. et al. Central resistin/TLR4 impairs
419–427 (2018). with brown adipose tissue transplant. Am. J. Physiol. adiponectin signaling, contributing to insulin and
101. Pflimlin, E. et al. Acute and repeated treatment with Endocrinol. Metab. 308, E1043–E1055 (2015). FGF21 resistance. Diabetes 65, 913–926 (2016).
5-PAHSA or 9-PAHSA isomers does not improve 126. Kloting, N. et al. Serum retinol-binding protein is more 149. Savage, D. B. et al. Resistin / Fizz3 expression in
glucose control in mice. Cell Metab. 28, 217–227 highly expressed in visceral than in subcutaneous relation to obesity and peroxisome proliferator-
(2018). adipose tissue and is a marker of intra-abdominal fat ­activated receptor-gamma action in humans. Diabetes
102. Syed, I. et al. Methodological issues in studying mass. Cell Metab. 6, 79–87 (2007). 50, 2199–2202 (2001).
PAHSA biology: masking PAHSA effects. Cell Metab. 127. Berry, D. C. et al. The STRA6 receptor is essential 150. Qatanani, M., Szwergold, N. R., Greaves, D. R.,
28, 543–546 (2018). for retinol-binding protein-induced insulin resistance Ahima, R. S. & Lazar, M. A. Macrophage-derived
103. Kuda, O. On the complexity of PAHSA research. but not for maintaining vitamin A homeostasis human resistin exacerbates adipose tissue
Cell Metab. 28, 541–542 (2018). in tissues other than the eye. J. Biol. Chem. 288, inflammation and insulin resistance in mice.
104. Lynes, M. D. et al. The cold-induced lipokine 24528–24539 (2013). J. Clin. Invest. 119, 531–539 (2009).
12,13-diHOME promotes fatty acid transport into 128. Moraes-Vieira, P. M. et al. Antigen presentation 151. Corre, J., Hebraud, B. & Bourin, P. Concise review:
brown adipose tissue. Nat. Med. 23, 631–637 and T-cell activation are critical for RBP4-induced growth differentiation factor 15 in pathology:
(2017). insulin resistance. Diabetes 65, 1317–1327 a clinical role? Stem Cells Transl Med. 2, 946–952
105. Stanford, K. I. et al. 12,13-diHOME: an (2016). (2013).
exercise-induced lipokine that increases skeletal 129. Thompson, S. J. et al. Hepatocytes are the principal 152. Ding, Q. et al. Identification of macrophage inhibitory
muscle fatty acid uptake. Cell Metab. 27, 1111–1120 source of circulating RBP4 in mice. Diabetes 66, cytokine-1 in adipose tissue and its secretion as an
(2018). 58–63 (2017). adipokine by human adipocytes. Endocrinology 150,
106. Huang-Doran, I., Zhang, C. Y. & Vidal-Puig, A. 130. Preitner, F., Mody, N., Graham, T. E., Peroni, O. D. & 1688–1696 (2009).
Extracellular vesicles: novel mediators of cell Kahn, B. B. Long-term Fenretinide treatment prevents 153. Lee, S. E. et al. Growth differentiation factor 15
communication in metabolic disease. Trends high-fat diet-induced obesity, insulin resistance, and mediates systemic glucose regulatory action of
Endocrinol. Metab. 28, 3–18 (2017). hepatic steatosis. Am. J. Physiol. Endocrinol. Metab. T-helper type 2 cytokines. Diabetes 66, 2774–2788
107. Chen, Y. et al. Exosomal microRNA miR-92a 297, E1420–E1429 (2009). (2017).
concentration in serum reflects human brown fat 131. Fedders, R. et al. Liver-secreted RBP4 does not impair 154. Hsu, J. Y. et al. Non-homeostatic body weight
activity. Nat. Commun. 7, 11420 (2016). glucose homeostasis in mice. J. Biol. Chem. 293, regulation through a brainstem-restricted receptor for
108. Thomou, T. et al. Adipose-derived circulating miRNAs 15269–15276 (2018). GDF15. Nature 550, 255–259 (2017).
regulate gene expression in other tissues. Nature 132. Lee, S. A., Yuen, J. J., Jiang, H., Kahn, B. B. & 155. Emmerson, P. J. et al. The metabolic effects of GDF15
542, 450–455 (2017). Blaner, W. S. Adipocyte-specific overexpression of are mediated by the orphan receptor GFRAL. Nat.
109. Ying, W. et al. Adipose tissue macrophage-derived retinol-binding protein 4 causes hepatic steatosis in Med. 23, 1215–1219 (2017).
exosomal miRNAs can modulate in vivo and in vitro mice. Hepatology 64, 1534–1546 (2016). 156. Mullican, S. E. et al. GFRAL is the receptor for GDF15
insulin sensitivity. Cell 171, 372–384 (2017). 133. Lu, J., Chatterjee, M., Schmid, H., Beck, S. & and the ligand promotes weight loss in mice and
110. BonDurant, L. D. & Potthoff, M. J. Fibroblast growth Gawaz, M. CXCL14 as an emerging immune and nonhuman primates. Nat. Med. 23, 1150–1157
factor 21: a versatile regulator of metabolic inflammatory modulator. J. Inflamm. (Lond.) 13, 1 (2017).
homeostasis. Annu. Rev. Nutr. 38, 173–196 (2018). (2016). 157. Xiong, Y. et al. Long-acting MIC-1/GDF15 molecules
111. Talukdar, S. et al. A long-acting FGF21 molecule, 134. Cereijo, R. et al. CXCL14, a brown adipokine that to treat obesity: evidence from mice to monkeys.
PF-05231023, decreases body weight and improves mediates brown-fat-to-macrophage communication in Sci. Transl Med. 9, eaan8732 (2017).
lipid profile in non-human primates and type 2 thermogenic adaptation. Cell Metab. 28, 750–763 This study showed that pharmacological
diabetic subjects. Cell Metab. 23, 427–440 (2016). (2018). administration of a GDF15 analogue reduces body
112. Schlein, C. et al. FGF21 lowers plasma triglycerides by This study describes the release of CXCL14 weight and improves metabolism in obese rodents
accelerating lipoprotein catabolism in white and brown by BAT into the circulation in response to and monkeys.
adipose tissues. Cell Metab. 23, 441–453 (2016). cold exposure, resulting in increased WAT 158. Fukuhara, A. et al. Visfatin: a protein secreted by
113. Hansen, J. S. et al. Glucagon-to-insulin ratio is pivotal browning and BAT activation via M2 macrophage visceral fat that mimics the effects of insulin. Science
for splanchnic regulation of FGF-21 in humans. recruitment. 307, 426–430 (2005).
Mol. Metab. 4, 551–560 (2015). 135. Pedersen, B. K. & Febbraio, M. A. Point: interleukin-6 159. Carbone, F. et al. Regulation and function of
114. Markan, K. R. et al. Circulating FGF21 is liver derived does have a beneficial role in insulin sensitivity and extracellular nicotinamide phosphoribosyltransferase/
and enhances glucose uptake during refeeding and glucose homeostasis. J. Appl. Physiol. 102, (814–816 visfatin. Compr. Physiol. 7, 603–621 (2017).
overfeeding. Diabetes 63, 4057–4063 (2014). (2007). 160. Stromsdorfer, K. L. et al. NAMPT-mediated NAD+
This study identified the liver as the source of 136. Schmidt-Arras, D. & Rose-John, S. IL-6 pathway in the biosynthesis in adipocytes regulates adipose tissue
circulating FGF21 and its insulin-sensitizing effects liver: from physiopathology to therapy. J. Hepatol. 64, function and multi-organ insulin sensitivity in mice.
in acute refeeding and overfeeding. 1403–1415 (2016). Cell Rep. 16, 1851–1860 (2016).
115. Liang, Q. et al. FGF21 maintains glucose homeostasis 137. Mohamed-Ali, V. et al. Subcutaneous adipose tissue 161. Xia, M. et al. Endothelial NLRP3 inflammasome
by mediating the cross talk between liver and brain releases interleukin-6, but not tumor necrosis activation and enhanced neointima formation in mice
during prolonged fasting. Diabetes 63, 4064–4075 factor-alpha, in vivo. J. Clin. Endocrinol. Metab. 82, by adipokine visfatin. Am. J. Pathol. 184, 1617–1628
(2014). 4196–4200 (1997). (2014).
116. BonDurant, L. D. et al. FGF21 regulates metabolism 138. Zhang, W. et al. Adipocyte lipolysis-stimulated 162. Kieswich, J. et al. Monomeric eNAMPT in the
through adipose-dependent and -independent interleukin-6 production requires sphingosine development of experimental diabetes in mice:
mechanisms. Cell Metab. 25, 935–944 (2017). kinase 1 activity. J. Biol. Chem. 289, 32178–32185 a potential target for type 2 diabetes treatment.
117. Hondares, E. et al. Thermogenic activation induces (2014). Diabetologia 59, 2477–2486 (2016).
FGF21 expression and release in brown adipose 139. Matsubara, T. et al. PGRN is a key adipokine 163. Wesener, D. A. et al. Recognition of microbial glycans
tissue. J. Biol. Chem. 286, 12983–12990 (2011). mediating high fat diet-induced insulin resistance and by human intelectin-1. Nat. Struct. Mol. Biol. 22,
118. Fisher, F. M. et al. FGF21 regulates PGC-1alpha and obesity through IL-6 in adipose tissue. Cell Metab. 15, 603–610 (2015).
browning of white adipose tissues in adaptive 38–50 (2012). 164. Yang, R. Z. et al. Identification of omentin as a novel
thermogenesis. Genes Dev. 26, 271–281 (2012). 140. Sabio, G. et al. A stress signaling pathway in adipose depot-specific adipokine in human adipose tissue:
119. Huang, Z. et al. The FGF21-CCL11 axis mediates tissue regulates hepatic insulin resistance. Science possible role in modulating insulin action. Am. J.
beiging of white adipose tissues by coupling 322, 1539–1543 (2008). Physiol. Endocrinol. Metab. 290, E1253–E1261
sympathetic nervous system to type 2 immunity. 141. Wunderlich, F. T. et al. Interleukin-6 signaling in (2006).
Cell Metab. 26, 493–508 (2017). liver-parenchymal cells suppresses hepatic 165. Watanabe, T., Watanabe-Kominato, K., Takahashi, Y.,
120. Keipert, S. et al. Genetic disruption of uncoupling inflammation and improves systemic insulin action. Kojima, M. & Watanabe, R. Adipose tissue-derived
protein 1 in mice renders brown adipose tissue a Cell Metab. 12, 237–249 (2010). omentin-1 function and regulation. Compr. Physiol. 7,
significant source of FGF21 secretion. Mol. Metab. 4, 142. Mauer, J. et al. Signaling by IL-6 promotes alternative 765–781 (2017).
537–542 (2015). activation of macrophages to limit endotoxemia and 166. Tan, B. K. et al. Omentin-1, a novel adipokine, is
121. Stanford, K. I. et al. Brown adipose tissue regulates obesity-associated resistance to insulin. Nat. Immunol. decreased in overweight insulin-resistant women with
glucose homeostasis and insulin sensitivity. J. Clin. 15, 423–430 (2014). polycystic ovary syndrome: ex vivo and in vivo
Invest. 123, 215–223 (2013). 143. Braune, J. et al. IL-6 regulates M2 polarization and regulation of omentin-1 by insulin and glucose.
122. Ruan, C. C. et al. A2A receptor activation attenuates local proliferation of adipose tissue macrophages in Diabetes 57, 801–808 (2008).
hypertensive cardiac remodeling via promoting brown obesity. J. Immunol. 198, 2927–2934 (2017). 167. Bluher, M. Vaspin in obesity and diabetes:
adipose tissue-derived FGF21. Cell Metab. 28, 144. Theurich, S. et al. IL-6/Stat3-dependent induction pathophysiological and clinical significance. Endocrine
476–489 (2018). of a distinct, obesity-associated NK cell subpopulation 41, 176–182 (2012).
This study provides evidence that hypertension- deteriorates energy and glucose homeostasis. 168. Kloting, N. et al. Vaspin gene expression in human
induced cardiac hypertrophy and fibrosis is Cell Metab. 26, 171–184 (2017). adipose tissue: association with obesity and type 2
counter-regulated by FGF21 induced in and 145. Xu, E. et al. Temporal and tissue-specific requirements diabetes. Biochem. Biophys. Res. Commun. 339,
secreted by BAT in this condition. for T-lymphocyte IL-6 signalling in obesity-associated 430–436 (2006).

NATuRe RevIeWS | EndOcRInOlOgy volume 15 | SEPTEMBER 2019 | 523


Reviews

169. Fain, J. N., Buehrer, B., Bahouth, S. W., Tichansky, D. S. 181. Kubota, T. et al. Pioglitazone ameliorates smooth 194. Sakamoto, Y. et al. Visceral adipose tissue-derived
& Madan, A. K. Comparison of messenger RNA muscle cell proliferation in cuff-induced neointimal serine protease inhibitor prevents the development of
distribution for 60 proteins in fat cells versus the formation by both adiponectin-dependent monocrotaline-induced pulmonary arterial
nonfat cells of human omental adipose tissue. and -independent pathways. Sci. Rep. 6, 34707 hypertension in rats. Pflugers Arch. 469, 1425–1432
Metabolism 57, 1005–1015 (2008). (2016). (2017).
170. Zieger, K. et al. Ablation of kallikrein 7 (KLK7) in 182. Zhou, M. et al. Rosiglitazone promotes fatty acyl CoA 195. O’Neill, S. M. et al. Targeting adipose tissue via systemic
adipose tissue ameliorates metabolic consequences of accumulation and excessive glycogen storage in livers gene therapy. Gene Ther. 21, 653–661 (2014).
high fat diet-induced obesity by counteracting adipose of mice without adiponectin. J. Hepatol. 53, 196. Aouadi, M. et al. Gene silencing in adipose tissue
tissue inflammation in vivo. Cell. Mol. Life Sci. 75, 1108–1116 (2010). macrophages regulates whole-body metabolism in
727–742 (2018). 183. Okada-Iwabu, M. et al. A small-molecule AdipoR obese mice. Proc. Natl Acad. Sci. USA 110,
171. Chou, S. H. et al. Leptin is an effective treatment for agonist for type 2 diabetes and short life in obesity. 8278–8283 (2013).
hypothalamic amenorrhea. Proc. Natl Acad. Sci. USA Nature 503, 493–499 (2013). This study describes a method to selectively target
108, 6585–6590 (2011). 184. Yamashita, T. et al. An orally-active adiponectin small interfering RNAs to macrophages in
172. Petersen, K. F. et al. Leptin reverses insulin resistance receptor agonist mitigates cutaneous fibrosis, epididymal WAT but not subcutaneous WAT or
and hepatic steatosis in patients with severe inflammation and microvascular pathology in a murine other organs.
lipodystrophy. J. Clin. Invest. 109, 1345–1350 model of systemic sclerosis. Sci. Rep. 8, 11843 (2018). 197. Chang, H. R., Kim, H. J., Xu, X. & Ferrante, A. W. Jr.
(2002). 185. Menzaghi, C. & Trischitta, V. The adiponectin paradox Macrophage and adipocyte IGF1 maintain adipose
173. Perry, R. J. et al. Mechanism for leptin’s acute for all-cause and cardiovascular mortality. Diabetes tissue homeostasis during metabolic stresses. Obesity
insulin-independent effect to reverse diabetic 67, 12–22 (2018). (Silver Spring) 24, 172–183 (2016).
ketoacidosis. J. Clin. Invest. 127, 657–669 (2017). 186. Aleksandrova, K., Mozaffarian, D. & Pischon, T. 198. Almuraikhy, S. et al. Interleukin-6 induces impairment
174. Oberlin, D. & Buettner, C. How does leptin restore Addressing the perfect storm: biomarkers in in human subcutaneous adipogenesis in
euglycemia in insulin-deficient diabetes? J. Clin. Invest. obesity and pathophysiology of cardiometabolic risk. obesity-associated insulin resistance. Diabetologia 59,
127, 450–453 (2017). Clin. Chem. 64, 142–153 (2018). 2406–2416 (2016).
175. Cui, H., Lopez, M. & Rahmouni, K. The cellular and 187. Zoi, I. et al. RANKL signaling and ErbB receptors in 199. Weiner, J. et al. Brown adipose tissue (BAT) specific
molecular bases of leptin and ghrelin resistance in breast carcinogenesis. Trends Mol. Med. 22, vaspin expression is increased after obesogenic diets
obesity. Nat. Rev. Endocrinol. 13, 338–351 (2017). 839–850 (2016). and cold exposure and linked to acute changes in
176. Levin, B. E. & Lutz, T. A. Amylin and leptin: 188. Kharitonenkov, A. et al. FGF-21 as a novel metabolic DNA-methylation. Mol. Metab. 6, 482–493 (2017).
co-regulators of energy homeostasis and neuronal regulator. J. Clin. Invest. 115, 1627–1635 (2005).
development. Trends Endocrinol. Metab. 28, 189. Sanyal, A. et al. Pegbelfermin (BMS-986036), Acknowledgements
153–164 (2017). a PEGylated fibroblast growth factor 21 analogue, The authors acknowledge the support of grants from the
177. Li, Z., Kelly, L., Heiman, M., Greengard, P. & in patients with non-alcoholic steatohepatitis: Deutsche Forschungsgemeinschaft (SCHE522/4-1),
Friedman, J. M. Hypothalamic amylin acts in concert a randomised, double-blind, placebo-controlled, Collaborative Research Center (SFB 1328) and Heisenberg
with leptin to regulate food intake. Cell Metab. 22, phase 2a trial. Lancet 392, 2705–2717 (2019). Professorship (HE3645/10-1).
1059–1067 (2015). 190. Kataoka, Y. et al. Omentin prevents myocardial
178. Roth, J. D. et al. Leptin responsiveness restored by ischemic injury through AMP-activated protein kinase- Author contributions
amylin agonism in diet-induced obesity: evidence and Akt-dependent mechanisms. J. Am. Coll. Cardiol. The authors contributed equally to all aspects of the article.
from nonclinical and clinical studies. Proc. Natl Acad. 63, 2722–2733 (2014).
Sci. USA 105, 7257–7262 (2008). 191. Matsuo, K. et al. Omentin functions to attenuate Competing interests
179. Lee, J. et al. Withaferin A is a leptin sensitizer with cardiac hypertrophic response. J. Mol. Cell Cardiol. The authors declare no competing interests.
strong antidiabetic properties in mice. Nat. Med. 22, 79, 195–202 (2015).
1023–1032 (2016). 192. Hiramatsu-Ito, M. et al. Omentin attenuates Peer review information
In this study the plant-derived molecule withaferin atherosclerotic lesion formation in apolipoprotein Nature Reviews Endocrinology thanks D. Langin and the other,
A was identified as a leptin sensitizer that reduces E-deficient mice. Cardiovasc. Res. 110, 107–117 anonymous, reviewer(s) for their contribution to the peer
body weight and improves glucose homeostasis of (2016). review of this work.
diet-induced obese mice. 193. Yuan, L. et al. Vaspin protects rats against myocardial
180. Saxena, N. K. & Anania, F. A. Adipocytokines and ischemia/reperfusion injury (MIRI) through the TLR4/ Publisher’s note
hepatic fibrosis. Trends Endocrinol. Metab. 26, NF-kappaB signaling pathway. Eur. J. Pharmacol. Springer Nature remains neutral with regard to jurisdictional
153–161 (2015). 835, 132–139 (2018). claims in published maps and institutional affiliations.

524 | SEPTEMBER 2019 | volume 15 www.nature.com/nrendo

You might also like