Anda di halaman 1dari 19

REVIEWS

Molecular pathways linking adipose


innervation to insulin action in obesity
and diabetes mellitus
Adilson Guilherme, Felipe Henriques   , Alexander H. Bedard    and Michael P. Czech*
Abstract | Adipose tissue comprises adipocytes and many other cell types that engage in dynamic
crosstalk in a highly innervated and vascularized tissue matrix. Although adipose tissue has been
studied for decades, it has been appreciated only in the past 5 years that extensive arborization
of nerve fibres has a dominant role in regulating the function of adipose tissue. This Review
summarizes the latest literature, which suggests that adipocytes signal to local sensory nerve
fibres in response to perturbations in lipolysis and lipogenesis. Such adipocyte signalling to the
central nervous system causes sympathetic output to distant adipose depots and potentially
other metabolic tissues to regulate systemic glucose homeostasis. Paracrine factors identified in
the past few years that mediate such adipocyte–neuron crosstalk are also reviewed. Similarly ,
immune cells and endothelial cells within adipose tissue communicate with local nerve fibres to
modulate neurotransmitter tone, blood flow , adipocyte differentiation and energy expenditure,
including adipose browning to produce heat. This understudied field of neurometabolism related
to adipose tissue biology has great potential to reveal new mechanistic insights and potential
therapeutic strategies for obesity and type 2 diabetes mellitus.

How fat is stored in mammals sparked vigorous scien- skeletal muscle, liver and brain10. Figure 1 (parts 1 and 2)
tific debate in the mid-​to-late 1800s, yielding conflicting illustrates several well-​studied pathways through which
concepts and false starts. Major schools of thought pro- this tissue crosstalk (Box 1) has been shown to operate.
posed that fat accumulated in unspecialized connective First, storage of lipids in white adipocytes, where these
tissue cells in the form of multilocular ‘mulberry’ cells or lipids are mostly derived from hydrolysis and intra­
that ‘wandering’ cells in the plasma accumulated fat and c­ellular re-​esterification of triglycerides in circulating
gathered in the connective tissue. Another view posited lipoproteins11–13, serves not only as a reservoir of calo­
the current concept that adipocytes occupy a special- ries for future use but also as a means to sequester
ized ‘glandular’ tissue, but as late as 1901, this hypo­thesis lipids away from peripheral tissues that are vulnerable
was the least appealing in a comprehensive review of to lipotoxicity that disrupts insulin’s actions on metabo-
the literature1. How surprising it would be to these early lism10,14,15. Thus, in obesity, fatty acids derived from die-
investigators that adipose tissue is indeed glandular in tary intake and white adipocyte lipolysis can increase
the true sense, functioning as an endocrine organ that hepatic gluco­neogenesis through the generation of the
can control systemic functions2–5, and that it secretes allosteric effector acetyl-​CoA16,17 and attenuate utiliza-
potent paracrine and autocrine factors that modify its tion of glucose in skeletal muscle by inhibiting glucose
glandular nature4,6,7. It would be even more startling for transport and metabolism18–21. These actions, combined
these investigators to learn that adipose tissue can be a with the dampening of pancreatic islet production of
major heat-​generating tissue, critical for many animals insulin, promote glucose intolerance, leading to type 2
to sustain body temperature during extreme cold expo- diabetes mellitus (T2DM). By contrast, high levels
Program in Molecular sure8,9. Viewed in this context, the multiple roles that of fatty acid oxidation, observed in the adipocytes of
Medicine, University of adipose tissue has in mammalian physiology are truly brown adipose tissue (BAT) and brown-​like beige
Massachusetts Medical
School, Worcester, MA, USA.
remarkable and unexpected. adipocytes (Box 1) in white adipose tissue (WAT) that
During the past couple of decades, it was also express UCP1 (Box 1) within the mitochondria, can also
*e-​mail: Michael.Czech@
umassmed.edu revealed that adipose tissue has a strong influence on decrease lipid accumulation in peripheral tissues and
https://doi.org/10.1038/ whole-​body glucose metabolism and lipid metabolism increase glucose tole­rance8. Thus, these main functions
s41574-019-0165-y through its effects on major tissues and organs such as of white adipocytes to store fat and brown adipocytes

Nature Reviews | Endocrinology


Reviews

Key points cells and local sensory and sympathetic nerve fibres
within the tissue, and on the potential to exploit these
• Adipocytes modulate whole-​body metabolism through secretion of endocrine and processes to develop strategies for future therapies
paracrine factors that modulate local immune cell cytokine secretion, endothelium targeting metabolic disease.
blood flow and neuronal signalling to the brain.
• Adipocytes, the endothelium and immune cells within adipose tissues secrete factors Adipose–sensory nerve fibre crosstalk
such as leptin, vascular endothelial growth factor (VEGF) and tumour necrosis factor Sensory innervation of adipose tissue
(TNF) that regulate local sensory nerve fibre functions.
Two modes of communication have been described
• Adipocyte lipid metabolism communicates with local sensory nerve fibres, sending whereby the metabolic status of adipose tissue is com-
signals to the central nervous system; conversely, sensory nerve fibres secrete factors
municated to the brain. The first is exemplified by the
such as calcitonin-​related gene peptide (CGRP) and substance P that might regulate
the metabolism of adipocytes and other adipose-​resident cells.
WATokine leptin, which is released from adipose tis-
sue into the circulation and acts on the hypothalamus
• Increased lipolysis in white adipose tissue in response to sympathetic activation
can cause sensory nerve fibres to regulate the metabolic activity of distant brown
to regulate appetite and systemic metabolism36,37. The
adipose tissue depots. discovery of leptin in 1994 definitively established a
• Extensive and dynamic signalling networks among the diverse cell types in adipose
functional role of adipocytes as endocrine cells37,38. The
tissue integrate and mediate communication through bioactive lipids to local sensory second mechanism of adipose to brain communication
nerve fibres and neurotrophic factors to sympathetic nerve fibres. is mediated through the sensory innervation of adipose
• Identifying factors within adipose tissues that regulate the function of sensory and tissue, which comprises an adipose–neuronal track
sympathetic nerve fibres might reveal therapeutic strategies for obesity and type 2 that can disseminate adipose tissue signals over long
diabetes mellitus. distances to the CNS39–41. Box 2 describes the historical
perspective of findings on the sympathetic and sensory
innervation of adipose tissue. Using the fluorescent
to oxidize fat allow adipose tissue to control systemic retro­grade tracer True Blue, sensory fibres in adipose
insulin sensitivity and susceptibility to metabolic dis- tissue were clearly observed in rats42. Moreover, immuno­
ease. Many reviews have extensively investigated the histochemical analysis of adipose tissue labelled
endocrine regulation of adipocyte lipid metabolism with antibodies against molecular markers selectively
in detail5,10,14,22. expressed in afferent neurons, such as calcitonin gene-​
Figure 1 (part 3) illustrates that white adipocytes related peptide (CGRP, which is produced by an alter-
modulate the accumulation and activities of immune native RNA splicing of the Calca gene transcripts and
cells within adipose tissues, for example, by attracting cleavage of inactive precursor protein (Box 1)), strongly
macrophages upon adipocyte death23 and by secreting supported the existence of adipose sensory innerva-
paracrine factors such as monocyte chemoattractant tion in hamsters and rats43,44. Subsequently, a series of
protein 1 (MCP1) and tumour necrosis factor (TNF) that elegant studies conducted by Bartness and colleagues
attract or activate macrophages, T and B lymphocytes showed that Siberian hamster adipose tissue is inner-
and other immune cells24–27. In turn, these cells secrete vated by sensory nerve fibres that convey signals to the
cytokines that can locally disrupt adipose lipid seques- brain40,45–48. In those studies, the H129 strain of herpes
tration10,14,28 and lipolysis29 or act systemically to inhibit simplex virus (HSV) type 1, an anterograde tract tracer,
insulin secretion or insulin action in other tissues30. was used to track routes between adipose tissue and the
Adipocytes also secrete factors (Fig. 1, part 4) termed brain through sensory nerves45,46.
adipokines (WATokines from WAT and BATokines Adipose tissue signals to the CNS through this affer-
from BAT) that can directly act on the local vasculature31 ent pathway can in turn trigger peripheral responses
(Fig. 1, part 5) or on distant tissues, with leptin being through sympathetic outflow, as has been reported
the prototypic such circulating factor that acts on the for WAT signalling to BAT in Siberian hamsters and
brain to suppress appetite and increase energy expendi- mice40,48,49. This intercommunication between adipose
ture32. Each of the above adipocyte-​mediated endocrine depots might be critical for energy balance and proper
and paracrine pathways can mediate systemic effects of control of systemic metabolism40,48,50. Accordingly,
adipose tissue on metabolic homeostasis. selective denervation of sensory fibres within WAT
A growing literature highlights an additional mode of suppresses sympathetic outflow and thermogenesis
adipose control over systemic metabolism — paracrine in BAT triggered by lipolysis in distant WAT depots in
signalling from adipocytes to localized nerve fibres Siberian hamsters48. Collectively, these results provide
(Box 2; Fig. 1, part 6). These adipocyte signals to local evidence that adipose sensory nerves have an impor-
nerve fibres can be transmitted to the central nervous tant role in detecting metabolic cues within adipose
system (CNS), which can then initiate output to other tissue and in conveying signals to the brain from such
tissues. Therefore, adipose-​initiated CNS stimulation cues, contributing to metabolic homeostasis (Fig. 2).
through local adipose afferent nerve fibres (Box 1) could
be a major modality of adipose control over systemic Adipose tissue signals to sensory nerves
glucose tolerance and insulin resistance. Although there Leptin. Progress in identifying the adipose-​derived
have been some previous reviews on this topic33–36, abun- molecular signals that regulate local sensory nerve
dant new relevant data have emerged over the past few fibres and ultimately modulate adipose–brain commu-
years (Box 2) and major new insights have been gained. nication has been encouraging (Fig. 3). Remarkably, the
The aim of this article is to critically review the most endocrine factor leptin can function in this capacity as
recent literature on signalling between adipose-​resident a local paracrine factor that modulates local afferent

www.nature.com/nrendo
Reviews

nerve fibres in adipose tissue51,52. Afferent fibres inner- of leptin was also shown to stimulate the sympathetic
vating WAT express the leptin receptor and are mark- nervous system (SNS) in skeletal muscle56. Altogether,
edly activated upon direct leptin injections into adipose these results suggest that leptin is one adipocyte factor
tissue in Siberian hamsters53. Additionally, selective that signals to local afferent nerve fibres, communi-
deletion of leptin receptor in vagal sensory neurons dis- cating the presence of increased fat stores to the brain.
rupts hypothalamic control of food intake and increases Thus, leptin acts both as a circulating factor to control
the propensity for obesity in mice54. Leptin-​stimulated whole-​body metabolism through hypothalamic regu­
afferent nerve fibres in WAT convey functional signals lation and as a paracrine factor to directly activate
to the CNS as they elicit enhanced sympathetic drive adipose afferent fibres that signal to the CNS. Impor­
(Box 1) into adipose tissue55. Central administration tantly, leptin release by adipocytes is itself acutely regu-
lated by local SNS activity, creating a highly integrated
signalling network36.
Efferent
Room temperature Cold temperature
nerve Fatty acids. Adipose-​tissue-derived fatty acids have also
been reported to be potent activators of sensory nerve
Noradrenaline fibres in adipose tissue40, which suggests that local affer-
β3-AR ent fibres sense adipose tissue lipolysis and communicate
Lipid
storage this metabolic flux to the CNS (Fig. 3). Importantly, in
1 Lipid 2 response to local fatty acid release from WAT, the brain
Lipid
oxidation increases sympathetic drive to BAT to enhance thermo-
release cAMP–PKA genesis in Siberian hamsters40. Other bioactive lipids
Lipids pathway
Cytokines such as arachidonic acid, eicosanoids and their derived
(immune Lipolysis Fatty lipids that are produced by adipocytes as well as other
cells) acid
Heat cells resident in adipose tissue, such as immune cells and
3 generation endothelial cells, have also been shown to act on local
AcCOA
sensory nerves40. Nonetheless, further investigation is
UCP1 necessary to more clearly determine the physiological
relevance of these bioactive lipids in the activation of
Mitochondrion adipose sensory nerves.
WATokines
White
White Brown or beige 4 Adipose-​derived neurotrophic factors
4
adipocyte
adipocyte adipocyte BATokines Another class of agents that is released by cells resi-
dent in adipose tissue that might regulate local afferent
fibres are neurotrophic factors (Box 1; Fig. 3a). These fac-
Blood vessel tors are bioactive molecules that control many aspects of
5 5
neuronal function in both the peripheral nervous system
and CNS. The neurotrophic molecules are essential for
6
neuronal development, regeneration, survival and main-
tenance57,58. Examples of adipose-​derived neurotrophic
Angiogenesis Angiogenesis
factors include neuregulin 4 (NRG4)59–61, brain-​derived
Afferent nerve neurotrophic factor (BDNF)62 and nerve growth fac-
tor (NGF)63. These proteins are powerful regulators of
Fig. 1 | Proposed mechanisms whereby adipose tissue controls systemic insulin proliferation, survival, migration and differentiation
sensitivity. Depicted are pathways in white adipocytes (left semicircle) and beige or of neurons and are implicated in several functions of
brown adipocytes (right semicircle) that have been proposed to affect whole-​body the systemic nervous system. These and other neuro-
insulin sensitivity and systemic metabolism in rodents and humans. At room temperature trophic peptides produced by adipose tissue might act
(22 °C) and above, most of the white adipose tissue (WAT) is composed of white
on local sensory nerves to expand innervation and/or
adipocytes (left), whereas at low temperatures (6 °C), brown and beige adipocytes appear
in WAT (right). White adipocytes promote fatty acid esterification into triglycerides for stimulate electrical activity62–65. Of particular interest,
storage, sequestering fat away from the liver and skeletal muscle to prevent lipotoxicity human adipocytes derived from adipocyte progenitors
(part 1). Sustained release of noradrenaline by adipose efferent nerves activates the that were differentiated and activated in vitro express
β3-adrenergic receptor (β3-AR) and induces the formation of beige adipocytes within neuroendocrine factors and the pro-​protein convertase
WAT. Beige adipocytes display increased mitochondrial density and high capacity for PCSK1 (also known as NEC1), variants of which are
fatty acid oxidation into acetyl-​CoA (AcCOA), which fuels heat production via strongly associated with obesity65. Results demonstrat-
mitochondrial UCP1 within the electron transport chain (part 2). White adipocytes ing that rodent sensory neurons express the receptors
upregulate resident immune cells in obesity , releasing cytokines into the circulation for NRG4, NGF and BDNF, such as the receptor tyrosine
(part 3). Secretion of white adipocyte-​derived bioactive molecules (denoted WATokines) protein kinases ERBB3 or ERBB4, TRKA (also known as
and beige or brown adipocyte-​derived factors (denoted BATokines) (part 4) might
high-​affinity NGF receptor) and TRKB (also known
modulate adipose vascularization (part 5) and activate local afferent nerve fibres
(part 6). A crosstalk between adipocytes and afferent nerve fibres (represented by the as BDNF/NT-3 growth factors receptor), respectively,
doubled-​headed arrows) might occur, as the afferent nerve can release neurotransmitters also support this notion66–68. Nonetheless, studies on
to communicate with adipose-​resident cells. In addition, these adipokines can be selective genetic deletion of these receptors in sensory
released into the circulation to affect distant tissues. BAT, brown adipose tissue; PKA , neurons will be necessary to determine whether they
protein kinase A. actually regulate adipose tissue sensory nerve functions

Nature Reviews | Endocrinology


Reviews

in vivo. A major caveat of this approach, however, is the a peptide known to stimulate angiogenesis and promote
lack of tools to ablate receptors selectively in adipose vascular sympathetic innervation and increase sensory
tissue sensory nerve fibres without perturbing sensory nerve density73–75. Findings published in the past 10 years
nerves in other tissues. have highlighted the role of adipose VEGFA in the con-
trol of adipose tissue function and in improving sys-
Vascular endothelial growth factor. The remarkably close temic energy metabolism and glucose tolerance through
anatomical and functional relationship between vascu- the enhancement of adipose vascularization 31,76,77.
larization and innervation of adipose tissue has been As sensory neurons express VEGF receptor (VEGFR)78,
established for a long time69–72. Morphological analy­sis sensory-​neuron-specific genetic deletion of VEGFR
revealed that fibres innervating adipose tissue juxtapose would be helpful to decipher whether VEGF acts through
the vasculature in rats69,70. Moreover, adipocytes secrete a sensory nerve VEGFR signalling pathway to enhance
vascular endothelial growth factor (VEGF)31 (Fig. 3a), adipose vascularization, sensory innervation and sys-
temic metabolism. In addition, the potential roles of the
individual isoforms of VEGF in this regard would be of
Box 1 | An explanation of specialist terms
interest and require further investigation.
β-Adrenergic receptor
The β-​adrenergic receptors belong to a class of G protein-​coupled receptors that are Cytokines. White adipocytes communicate with immune
activated by catecholamines. cells that are resident in the adipose tissue, such as macro­
Afferent nerves phages and lymphocytes, to modulate cytokine produc-
These nerves consist of sensory nerve fibres that can transmit information from the tion and thus regulate the levels of adipose inflammation,
periphery to the central nervous system. as occurs in humans and rodents with obesity25,79,80.
Beige adipocytes Cytokines secreted from macrophages, such as TNF,
These UCP1-positive, thermogenic adipocytes appear within white adipose tissue IL-1β and IL-6, can act directly on the sensory neuron
(WAT) upon stimulus. These cells are often called inducible brown, beige or brite (brown itself81–83 or indirectly through their pro-​inflammatory
in white) adipocytes. effects within the adipose tissue microenvironment
Browning of WAT (Fig. 3a). By contrast, anti-​inflammatory cytokines, such
This process involves the formation of beige adipocytes and/or the conversion of white as IL-17A and adenosine, which originate from alter­
adipocytes into beige adipocytes within WAT that occur in response to cold or other natively activated macrophages, regulatory T (Treg) cells
stimuli. Through this process, expression of UCP1 protein and the thermogenic capacity and T helper cells, can also interact directly with sen-
of WAT are enhanced. sory nerve terminals or indirectly through modulation
calcitonin gene-​related peptide of the adipose microenvironment to elicit neuronal
Calcitonin gene-​related peptide (CGRP) is a 37-amino-​acid neuropeptide, produced as responses84,85. As such, the inflammatory status of adi-
a consequence of alternative RNA processing of the calcitonin gene (CALCA) and pose tissue is likely to be highly influential on afferent
primarily associated with sensory nerve fibres. signalling to the CNS. Altogether, these observations
crosstalk suggest that many bioactive molecules that are derived
In this Review, we refer to the intercommunication between adipose-​resident cells and from adipose tissue function as paracrine factors to regu-
the local neuronal fibres as crosstalk, which is determined by the interactions between late local sensory nerves and that additional such factors
secreted molecules from one cell type with the related receptor or receptors in a are still to be discovered.
distinct cell type.
Efferent nerves Sensory nerve factors
These nerves consist of sympathetic nerve fibres that travel away from the central Calcitonin gene-​related peptide. Whereas adipose-​
nervous system to innervate adipose tissue and other peripheral tissues. resident cells secrete factors that act on local sensory
Hyperinsulinaemia nerve fibres, stimulated sensory nerves might also
A state of supraphysiological circulating levels of insulin. It is closely associated with release factors that modulate adipose tissue function
glucose intolerance, obesity, hypertension and dyslipidaemia. in a way that affects whole-​body metabolism (Fig. 3b).
Macrophage polarization This phenomenon, known as the axon reflex, enables
The shift in macrophage phenotypic profile between pro-​inflammatory M1 subtypes afferent fibres to signal directly to the peripheral tissue
and anti-​inflammatory M2 subtypes depending on external cues. that they innervate, circumventing central signal inte-
neurogenic inflammation gration and response86. An example is CGRP, a member
Local inflammation generated by signals originating from nerve fibres. of the calcitonin family of peptides, which is synthesized
neuropathy and released from nerves in the CNS as well as from
The damage or destruction of nerves. peripheral nerve fibres87. CGRP expression seems to
be concentrated in sensory afferents that innervate the
neurotrophic factors
vasculature87, which suggests that the endothelium is a
These are molecules that regulate growth, morphological plasticity and the survival
potential of neurons.
primary target for CGRP released by sensory nerves.
Accordingly, CGRP acts as a potent vasodilator when
Sympathetic drive it binds to its cognitive calcitonin receptor-​like recep-
Sympathetic nerve activity.
tor (CALCRL; also known as CGRP type 1 receptor) in
UcP1 endothelial cells88. The activation of CALCRL signalling
A mitochondrial protein found in beige and brown adipocytes that is involved in has also been reported to promote immune cell adhesion
thermogenesis. It uncouples mitochondrial respiration from ATP production, and migration through the endothelium and to modu-
converting energy of membrane electrical potential into heat.
late tissue inflammation89. In this regard, it is conceivable

www.nature.com/nrendo
Reviews

Box 2 | Historical perspective of adipose tissue innervation from sensory nerve fibres within adipose tissue directly
modu­lates adipocyte metabolism is suggested from a
It has long been known that adipose tissue is innervated, but the extent of adipose study revealing lipolytic responses to CGRP adminis-
innervation and its functional importance has become increasingly apparent in the past tered systemically92. Thus, further exploration of CGRP
few years. Sympathetic nerve fibres within adipose tissue were revealed by the mid- as a paracrine factor affecting adipocytes within adipose
1960s71,239, but evidence of sensory innervation of adipose tissue was not reported until
tissue is warranted.
>20 years later42,240. Beginning in the 1990s, a growing number of publications appeared
on the role of adipose tissue innervation in adipocyte function. The Bartness laboratory,
in particular, generated a deep literature on this topic, and adipose tissue biology Substance P. Substance P is a neuropeptide released by
experienced a renaissance as well, generating the realization that it is a highly complex peripheral sensory nerves that, similar to CGRP, acts as
endocrine organ with extensive influence over systemic homeostasis. Key discoveries a potent vasodilator and immune cell regulator93 (Fig. 3b).
in this field have included the finding that fasting increases adipocyte lipolysis primarily Substance P exerts its biological activity via the high-​
as a result of local release of catecholamines through enhanced adipose sympathetic affinity neurokinin 1 receptor (NK1R; also known as
activity rather than through increased circulating levels of catecholamines133,240. substance P receptor) in endothelial and immune cells94.
Similarly, it was discovered that the antilipolytic effect of insulin is also in part mediated Interestingly, substance P stimulates immune cells to
through inhibition of sympathetic activity at the level of the central nervous system express several pro-​inflammatory cytokines, such as
(CNS)122,134. The discovery of leptin in 1994 revealed the capacity of adipose tissue to
IL-6 and TNF95, which suggests a role for this neuro-
communicate with the CNS and other distant organs through the circulation, and the
discovery of many additional adipokines with various potential activities has continued peptide as a mediator of adipose tissue inflammation
to reinforce this paradigm32. in obesity and metabolic dysfunction. Selective genetic
The graph, generated from PubMed searches of the terms listed, shows the rapidly deletion of NKR1 receptors expressed in endothelial
expanding discovery of neurotrophic factors emitted by adipose tissue that could directly or immune cells in the adipose tissue will be necessary
interact with local nerve fibres and other cell types within the tissue. Such adipocyte– to definitively determine whether substance P–NKR1
nerve crosstalk has added to our knowledge of the complexity of adipose tissue and its signalling modulates adipose tissue functions and
relationship to whole-​body metabolism and has created a new perspective on the basis whole-​body metabolism.
of metabolic disorders such as diabetes mellitus, obesity and cachexia. Targeting these
disorders through the neuro–adipose connection offers new strategies for future Sensory dysregulation in obesity
therapeutic interventions. The number of publications was identified in PubMed and is
A hallmark of adipose tissue dysfunction in obese ani-
expressed as the total number of results for the specific keywords “adipose tissue
innervation” (blue bars) and “adipose tissue and neurotrophic factors” (red bars). mal models and humans with obesity is a low-​grade
chronic inflammatory state10,79,96. Immune cell infil-
50
tration, macrophage proliferation and increased pro-​
Adipose tissue innervation
Adipose tissue and neurotrophic factors inflammatory cytokine production are noted in adipose
tissue in rodent and human obesity25,79. As inflammatory
40
1964 1988 1994 mediators trigger neuritis and disrupt sensory neuron
Number of papers (PubMed)

BAT efferent BAT afferent Discovery activity81,97, the increased production of cytokines in
innervation innervation of leptin
30
adipose tissue might impair the ability of local sensory
1970 1987 nerves to convey information from the adipose tissue
WAT efferent WAT afferent
innervation innervation to the brain. Indeed, sensory nerve dysfunctions in the
20 initial stages of glucose intolerance in patients with
obesity have been reported98. Thus, obesity-​associated
adipose dysfunction and chronic inflammation that
10 disrupts local afferent signalling to the CNS might have
adverse consequences for adipose tissue functions and
whole-​body metabolism. Studies designed to test this
0 hypothesis will be of high interest.
Sensory nerve fibres in adipose tissue might also
57

65

69

73

77

81

85

89

93

97

01

05

09

13

17
19

19

19

19

19

19

19

19

19

19

20

20

20

20

20

contribute to the adipose tissue inflammation in obe-


BAT, brown adipose tissue; WAT, white adipose tissue. sity. Hypothetically, the adipose tissue expansion and
increased release of leptin and free fatty acids in humans
that CGRP released by adipose tissue sensory nerves and rodents with obesity could cause hyperactivation of
has a key role in adipose vascular homeostasis and local sensory nerve fibres and consequent release of vaso­
inflammation and thus systemic metabolism. active and pro-inflammatory neurotransmitters, such
Studies published in the past decade have shown as CGRP and substance P. This process has been well
that genetic deletion of the Calca gene causes increased studied in other peripheral tissues and is known as neuro­
energy expenditure and insulin sensitivity with resist- genic inflammation99,100 (Box 1). This type of inflam-
ance to diet-​induced obesity without changes in food mation occurs when activated afferent nerves release
intake 90,91. Interestingly, these effects were accom- inflammatory mediators, triggering tissue inflamma-
panied by evidence of increased SNS activity, which tion99,100. Importantly, neurogenic inflammation also
is consistent with the concept that CGRP dampens seems to have a role in neuritis and nerve damage, which
activity of sympathetic nerve fibres. Whether these are pathologies often seen in patients with obesity, type 1
phenotypes noted in mice lacking the Calca gene are diabetes mellitus and T2DM101,102. Therefore, an excit-
due to changes in adipose tissue vascular function and ing hypothesis to test is whether sensory neurogenic
levels of adipose tissue inflammation has not yet been inflammation contributes to adipose inflammation and
investigated. However, the concept that CGRP released dysfunction in obesity.

Nature Reviews | Endocrinology


Reviews

a Afferent pathway b CNS c Efferent pathway d Systemic effects


(sensory) (sympathetic)

BAT

Sensory
nerve ?
Liver
Sympathetic
nerve

Muscle

Signalling
Signalling factors
factors
?
WAT Pancreas

Fig. 2 | Adipose signalling to local nerve fibres regulates systemic metabolism. a | Sensory nerves (dark blue arrows)
relay information from the white adipose tissue (WAT) microenvironment to the central nervous system (CNS). b | The CNS
integrates adipose tissue signals to orchestrate a response to the adipose tissue microenvironment. The CNS conveys its
response via sympathetic outflow (light blue arrows) back into the periphery. c | The sympathetic nerve innervating WAT
releases signalling factors that influence the adipose tissue microenvironment. d | The autonomic nervous system also
affects other metabolic organs to promote whole-​body homeostasis. Whether adipose metabolic cues are conveyed to
the CNS to control sympathetic outflow into liver, muscle and pancreas (light blue arrows) is still unknown. Thus, the
adipose tissue microenvironment might have a role in regulating systemic metabolism through signalling to local nerve
fibres. The depicted cartoon illustrates a general concept. The scale in which the diagrams were drawn is not anatomically
accurate. BAT, brown adipose tissue.

Central integration of adipose signals afferent nerves traversed up the spinal cord, localizing
One key area of focus introduced by the findings that in all areas of the brain — the hindbrain, midbrain and
adipose tissue depots are innervated by sensory fibres forebrain. Of these areas, the forebrain is highly relevant
is determining the destination and central function of to systemic metabolism, as it contains the hypothalamus.
these afferent fibres. Analyses conducted by the Bartness
group applying anterograde tract tracing with HSV Essential role of the hypothalamus
H129 demonstrated that sensory innervation of adi- The hypothalamus (Fig. 4) is essential for regulation of
pose tissue arises from dorsal root ganglion neurons45,46. thermogenesis103,104, energy balance and systemic meta­
This neuronal tracing method also revealed extensive bolism105–107, integrating signals from peripheral organs
projections of the adipose afferent network as well as and tissues to generate proper physiological responses.
surprising results regarding the anatomy of afferent In addition to neuroendocrine signalling to the pitui-
fibres innervating different adipose tissue depots. For tary108, the hypothalamus relies on neuronal circuits and
instance, subcutaneous inguinal WAT (iWAT), the WAT connections to communicate directly with peripheral
depot most susceptible to browning (Box 1), seems to organs and tissues. One example of such a neuronal cir­
be entirely innervated by spinal afferents that typically cuit is the adipose–hypothalamus–adipose neuronal
associate with somatosensory information. Moreover, no circuit40,41. As shown in Fig. 4a, the pseudounipolar affer-
signal arising from the nodose ganglion, which is typi- ent sensory fibres that innervate WAT depots arise from
cally associated with visceral afferents, was detected in the dorsal root ganglion proximal to the spinal cord. The
iWAT. Thus, this distinct sensory innervation in iWAT dorsal root ganglion, which also projects to the brain via
probably contributes to its increased ability to promptly the dorsal horn of the spinal cord, relays sensory infor-
respond to changes in physiological conditions such mation from the periphery to the CNS for integration
as cold exposure compared with other adipose depots. (Fig. 4a). Then, primarily through this hypothalamic
Importantly, these analyses also revealed that these integration, signals can be conveyed as sympathetic

www.nature.com/nrendo
Reviews

outflow to adipose tissue and peripheral organs via the depots of WAT. Dual pseudorabies virus tracing of mes-
intermediolateral nucleus of the spinal cord (Fig. 4a). enteric WAT (mWAT) and iWAT to reveal the sympa-
Subsequent studies utilizing both anterograde HSV thetic pathways innervating both tissue depots showed
H129 tracing and retrograde pseudorabies virus trac- that iWAT had considerably more single-​labelled neu-
ing from white and brown adipose depots in Siberian rons within the brain than mWAT110. However, areas
hamsters revealed areas of colocalization within the more typically associated with sympathetic regulation of
hypothalamus, suggesting the existence of a reflex cir- metabolism, such as the paraventricular hypothalamic
cuit that converts adipose sensory cues into sympathetic nucleus, lacked such a striking difference in labelling.
outflow back into adipose tissue39,109. Corroborating this This finding suggests that the differences observed in
notion, in an extensive screening of central HSV H129 metabolic phenotypes associated with subcutaneous
and pseudorabies virus colocalization from iWAT injec- versus visceral adiposity might have a neuronal compo-
tions, Bartness and colleagues revealed a high degree of nent41. If the neural origins of sympathetic innervation
doubly labelled neurons (upwards of 75%), suggesting vary between adipose depots, a hardwired differential
an extensive sensory-​sympathetic iWAT feedback sys- sympathetic drive to each tissue might exist, further
tem in Siberian hamsters47. However, an important point suggesting specialized, depot-​specific roles.
to consider related to these findings is the caveats asso- An early study (published in 1998) also showed less
ciated with the use of HSV H129. In particular, double pseudorabies virus labelling within the suprachias-
labelling of afferent and efferent nerve fibres (Box 1) is matic, arcuate and dorsomedial hypothalamic nuclei
possible, making the interpretation of the above results following injections into epididymal WAT (eWAT)
less clear. Moreover, as the HSV H129 tracing experi­ viral injections than after injections into iWAT111. One
ments were conducted in hamsters, replication from should note that when dual-​tract tracing is performed,
these results in other animal species, such as mice, will one virus can interfere with the infection of the same
be necessary to extend these findings. neuron by the second, leading to an underestimation
An additional remarkable finding was the obser- of dually labelled neurons. These critical studies indi-
vation of differential sympathetic outflows to various cate that adipose sensory and sympathetic nerves are

a
Afferent pathway
Adipose-tissue-secreted CGRP and substance P (sensory)
factors reported to released from sensory
affect afferent nerves nerve terminals
• Leptin Molecular mediators Brain
• NRG4 released from adipose-
• NGF tissue-resident cells
• BDNF
• FFAs
• AA
• EPA
b
Treg
• PGE2 Afferent-nerve-secreted
cell factors reported to
• VEGF White
adipocyte affect adipose tissue
• TNF • CGRP
• IL-1β • Substance P
• IL-6
• IL-17A Beige
• Adenosine adipocyte

Blood
vessel Macrophage

Fig. 3 | Adipose tissue–sensory nerve crosstalk. a | Adipose-​tissue-resident cells release molecular mediators
(represented by yellow circles) that can act on the afferent neuronal pathway and invoke a central response to the
tissue microenvironment. The principal component of adipose tissue, the adipocyte, can release various neuroactive
and neurotrophic peptides, such as leptin, neuregulin 4 (NRG4), nerve growth factor (NGF), brain-​derived neurotrophic
factor (BDNF), free fatty acids (FFAs), arachidonic acid (AA) and eicosanoids, such as eicosapentaenoic acid (EPA) and
prostaglandin E2 (PGE2), that act on surrounding cells, including sensory nerve fibres. Adipocytes and the blood vessels
within the adipose tissue secrete vascular endothelial growth factor (VEGF), which can promote nerve sprouting and
sensory hypersensitization upon interaction with the VEGF receptor (VEGFR) family. Cytokines secreted from
macrophages, such as tumour necrosis factor (TNF), IL-1β and IL-6, might act directly on the sensory nerve itself or
indirectly through their pro-​inflammatory effects within the adipose tissue microenvironment. Anti-​inflammatory
cytokines, such as IL-17A and adenosine, originating from alternatively activated macrophages and various lymphocytes,
including regulatory T (Treg) cells, can act in a similar way. b | Conversely , signalling from the sensory nerve terminals to cells
within adipose tissue has the potential to modulate adipocyte functions. Upon stimulation, the sensory nerve can release
calcitonin gene-​related peptide (CGRP) and substance P (represented by blue circles) into the innervated tissue that
modulates the microenvironment surrounding the sensory nerve.

Nature Reviews | Endocrinology


Reviews

a Peripheral signalling to the b


Central nervous system Hypothalamus
central nervous system
Inflamed
Lean Obesity

DR
LH LH

G
PVN PVN

IML POA POA


WAT DMH DMH

Spinal cord SCN VMH VMH SCN

Sympathetic
chain ARC ARC
Microglia
3V
Astrocyte

RPa RVLM

IML

Fig. 4 | central integration of adipose signals and obesity-​mediated dysregulation. a | Afferent sensory nerve fibres
innervating white adipose tissue (WAT) depots arise from the dorsal root ganglia (DRG) proximal to the spinal cord.
The DRG also projects to the brain via the dorsal horn of the spinal cord, relaying sensory information from the periphery
to the central nervous system for integration. b | The hypothalamus is a primary area for metabolic regulation in the
central nervous system, influencing thermogenesis and food intake as well as other critical homeostatic functions
throughout the body. Projections into the preoptic area (POA), as well as resident temperature-​sensitive neurons,
relay critical thermoregulatory information to the dorsomedial hypothalamic nucleus (DMH), a core component of the
orexinergic system and thermoregulatory function of the hypothalamus. The paraventricular hypothalamus (PVN), which
is proximal to the third ventricle (3V), is involved in food intake, thermoregulation and neuroendocrine functions through
projections to the pituitary. The arcuate nucleus (ARC), along with the ventromedial nucleus of the hypothalamus (VMH)
and lateral hypothalamus (LH), are also involved in appetitive behaviour and food reward. The suprachiasmatic nucleus
(SCN) is a critical area for regulating circadian rhythms. All of these centres have direct or indirect roles in influencing
hypothalamic thermogenic regulation. Hypothalamic inflammation has been linked to metabolic dysregulation and
obesity-​related insulin resistance through excessive gliosis, leading to neuronal damage, particularly noted within the
ARC. The hypothalamus sends sympathetic projections to the periphery either directly through the intermediolateral
nucleus of the spinal cord (IML), or via relay through the raphe pallidus nucleus (RPa) or the rostral ventrolateral medulla
(RVLM) to the IML.

connected and have the potential for communication insulin signalling in peripheral tissues and systemic
via hypothalamic relay, as well as via areas of the hind- metabolism. Insulin was shown to engage with insulin
brain and midbrain. Additionally, divergences in these receptors in two neuronal populations: the cocaine-​
pathways between WAT depots have emerged as pro- related and amphetamine-​related transcript protein–
vocative areas to probe when addressing the phenotypic proopiomelanocortin (CART–POMC) neurons and the
disparity contributed by visceral and subcutaneous adi- neuropeptide Y–agouti-​related peptide (NPY–AGRP)
posity. Although these adipose–hypothalamus–adipose neurons present within the arcuate nucleus of the hypo-
neuronal signalling axes exist as potentially important thalamus116,117. Insulin signalling in these neurons affects
networks in metabolic communication, traditional neuronal excitability and peptide expression and stim-
regulators of sympathetic drive, such as central lep- ulates the appetite-​suppressing population of POMC
tin and insulin signalling, remain prominent modes neurons, which in turn inhibits the appetite-​promoting
of metabolic signalling within the hypothalamus. It is population of AGRP neurons116,118,119. Importantly, insu-
important to emphasize the essential role of central lep- lin receptor signalling in POMC and AGRP seems to be
tin signalling in the regulation of regional sympathetic essential to regulate energy balance and systemic metab-
nerve activity, energy expenditure and metabolic homeo­ olism, as genetic disruption of components in the insu-
stasis. Excellent reviews discussing the contribution of lin receptor signalling pathway in those neurons alters
hypothalamic leptin signalling on SNS activation and systemic metabolism120,121. For instance, insulin recep-
glucose systemic metabolism have been published in the tor deletion in POMC neurons abolishes the ability of
past few years36,107,112–115. insulin to suppress adipose lipolysis, while preserving its
ability to suppress hepatic glucose production, and led to
Hypothalamic insulin signalling the development of hepatic steatosis in mice fed a high-​
The functional relevance of adipose–hypothalamic neu- fat diet122. Furthermore, deletion of insulin receptors
ronal circuitry for adipose thermogenesis and therefore in AGRP neurons impaired insulin’s ability to suppress
systemic metabolism was also demonstrated in experi- hepatic glucose production, whereas its action to inhibit
ments aimed at investigating the role of hypothalamic lipolysis was preserved122.

www.nature.com/nrendo
Reviews

The observations discussed in this section raise an triacylglycerol in white adipocytes during fasting con-
important question regarding the relevance of central ditions133 (Fig. 5). By contrast, the secretion of adrenaline
integration for metabolic homeostasis. Results support- by the adrenal glands does not seem to be required for
ing an essential role of the CNS and hypothalamus in adipocyte lipolysis during fasting133, which supports the
controlling whole-​body metabolism are well established. notion that adipose sympathetic drive is indispensable
In one such study, chronic decerebrated rats were shown for mobilization of free fatty acids and their use as fuel
to be hyperinsulinaemic (Box 1), hyperadiponectinaemic in other tissues. The importance of adipose sympathetic
and hyperleptinaemic compared with their control coun- drive for the proper control of adipose tissue lipolysis is
terparts123. The presence of the forebrain was shown to be also illustrated by the experiments from Buettner and
critical for maintaining normal circulating levels of insu- colleagues in which central insulin administration sup-
lin, body temperature, energy expenditure and a healthy presses lipolysis in peripheral adipocytes134. Thus, insu-
balance of lean and fat mass123. Whether these findings lin acts not only cell autonomously in adipocytes but also
result from a disrupted neuroendocrine axis, disrupted in the hypothalamus to attenuate lipolysis. The actions in
direct sympathetic outflow or a combination of the two the hypothalamus occur through insulin-​mediated inhi-
should be further investigated. Altogether, these results bition of sympathetic activity in adipose tissue through
demonstrate the importance of the central integration of unknown mechanisms134. Collectively, these observa-
peripheral signals and the hypothalamic regulation tions provide strong evidence that major physiological
of adipose sympathetic drive and systemic metabolism. pathways that mediate both stimulation and inhibition
of adipocyte lipolysis are driven by noradrenaline and
Central dysregulation insulin regulation of sympathetic neurons. Thus, brain
Obesity causes notable morphological changes within to adipocyte communication through sympathetic
the hypothalamus in a mouse model124–128. In particu- innervation of adipose tissue seems to be pivotal for
lar, it enhances inflammation and microglial expan- homeostatic control of whole-​body metabolism50.
sion124–128 (Fig. 4b). These observations were extended Sympathetic nerve fibres are readily detected within
to individuals with obesity who displayed increased the adipose parenchyma, and it has been suggested that
inflammation and gliosis throughout the mediobasal a cold stimulus triggers sympathetic expansion as well as
hypothalamus, along with functional impairment124,129. increased SNS activity in adipose tissue135,136. However,
Moreover, results describing mediobasal hypothalamus using a whole-​tissue clearing method that permits visu-
gliosis linked to obesity and insulin resistance in humans alization of fluorescently labelled structures in an entire
have been published in the past few years129,130. Thus, adipose depot, Paul Cohen’s group quantified the degree
persistent hypothalamic inflammation positively corre- of arborization of anti-​tyrosine hydroxylase antibody-​
lates with metabolic dysfunction in animal models of positive structures in iWAT and found (using light sheet
obesity and humans with obesity. To elucidate whether microscopy) no change in response to cold exposure137.
hypothalamic inflammation is implicated in metabolic This finding appears to be contradictory to the results
dysfunction, studies from the past few years have inves- previously described here136 and to western blot analyses
tigated whether targeting hypothalamic inflammation that demonstrate an increase in iWAT tyrosine hydroxy­
would improve systemic metabolism. In one study, evi- lase protein levels when mice are housed in the cold137.
dence suggested that the NF-​κB pathway is necessary Taken together, the data suggest that tyrosine hydroxy-
for hypothalamic microgliosis and metabolic dysfunc- lase expression is increased by cold exposure in nerve
tion in obesity in mice131. Through either pharmacolog- fibres that are already present in iWAT (that is, cold-​
ical depletion of microglia or inhibition of the NF-​κB induced sympathetic stimulation without structural
pathway in these cells, hypothalamic inflammation was remodelling)137. This issue will be of future interest as
prevented and a normal systemic energy balance was other laboratories use light sheet microscopy to explore
preserved131. Overall, these results strongly support nerve fibres within adipose tissue depots.
the notion that hypothalamic inflammation in obesity The increased sympathetic innervation within
contributes to metabolic dysfunction and energy imbal- WAT and BAT parenchyma is critical for the increased
ance. Moreover, persistent hypothalamic inflammation expression of genes encoding protein products involved
can affect central integration and systemic metabolism, in thermogenesis in adipose tissue in response to cold
disrupting the adipose afferent reflex by uncoupling exposure135,136,138. Accordingly, selective sympathetic
the adipose microenvironment from the central relay denervation of WAT and BAT disrupts cold-​induced
through sympathetic outflow into the periphery. thermogenesis136,138,139. The importance of adipose sym-
pathetic drive for thermogenesis and systemic metab-
Adipose–sympathetic nerve fibre crosstalk olism was also consolidated in a report showing that
Sympathetic innervation of adipose regional sympathectomy impairs adipose thermogenesis
Immunohistochemical analysis of selective sympathetic and leaves mice susceptible to obesity50.
neuron markers, such as the catecholamine biosynthetic
enzyme tyrosine hydroxylase, revealed sympathetic Sympathetic nerve factors
nerve fibres in adipose tissue in close anatomical asso- Efferent sympathetic nerve fibres regulate adipose
ciation with the vasculature, similar to what is observed tissue functions through the secretion of the neuro-
with adipose afferent nerves69,132. Sympathetic inner- transmitters noradrenaline, NPY and ATP (Fig.  5a).
vation of adipose tissue and release of noradrenaline These sympathetic-​derived factors are present within
from these nerve fibres are also essential for lipolysis of the nerve fibres and are released upon the generation

Nature Reviews | Endocrinology


Reviews

Efferent pathway
Molecular mediators (sympathetic)
released from adipose-
tissue-resident cells
Brain
Noradrenaline,
NPY or ATP
a b
Efferent-nerve-secreted Adipose-tissue-secreted
factors reported to factors reported to
affect adipose tissue affect efferent nerves
• Noradrenaline Treg
cell • NRG4
• NPY White • NEGR1
• ATP adipocyte • NGF
• BDNF
• FFAs
• Endocannabinoids
Beige • VEGF
adipocyte • TNF
• IL-1β
Blood • IL-6
vessel Macrophage • IL-10
• GDF15
• Adenosine

Fig. 5 | Adipose tissue–sympathetic nerve crosstalk. a | Efferent nerve fibres are known to regulate adipose tissue
functions through secretion of bioactive factors (represented by blue circles). Among them are catecholamine
(noradrenaline), neuropeptide Y (NPY) and ATP. The central nervous system stimulates sympathetic outflow to adipose
tissue, triggering the secretion of noradrenaline, NPY or ATP. These sympathetic-​derived secreted factors, through the
activation of their respective receptors, affect not only adipocytes but also other adipose-​resident cells, such as
endothelial cells, macrophages and lymphocytes. b | In turn, the stimulated adipose-​resident cells also produce a number
of secreted bioactive factors (represented by the yellow circles) that communicate with adipose sympathetic fibres. For
instance, multilocular beige adipocytes (the formation of which is induced via sympathetic noradrenaline and/or
adenosine molecules) produce the neurotrophic factor neuregulin 4 (NRG4), which is known to promote neurite
outgrowth. Additionally , white adipocytes synthesize several factors with neurotrophic activity that might enhance
sympathetic innervation of adipose tissue. Among these factors are neuronal growth regulator 1 (NEGR1), nerve growth
factor (NGF), brain-​derived neurotrophic factor (BDNF), free fatty acids (FFAs) and endocannabinoids. The vascular
endothelial growth factor (VEGF) secreted by the endothelium and adipocytes elicits sympathetic innervation in adipose
tissue. Adipose-​resident immune cells, such as macrophages and lymphocytes, secrete cytokines, tumour necrosis factor
(TNF) and IL-1β as well as factors demonstrated to affect neurite outgrowth and possibly adipose sympathetic innervation,
such as IL-6, IL-10 and growth differentiation factor 15 (GDF15). Treg, regulatory T cell.

of an action potential. Reports suggesting sympathetic Noradrenaline. The neurotransmitter noradrenaline and
co-​transmission and coordinated actions of noradren- its role in adipose metabolic processes have been well
aline and ATP electrically released from nerve endings studied for many years and its actions within adipose
in blood vessels have been published over the past three tissue have been reviewed in the past few years148,149.
decades140–142. Importantly, although results depicting Upon CNS stimulus, sympathetic nerve fibres release
sympathetic co-​transmission by the adipose nerve end- noradrenaline within adipose tissue that, upon binding
ings are not abundant, evidence supports the existence of to adrenergic receptors, modulates several metabolic
this process in adipose nerve terminals143. Accordingly, processes, including lipolysis, thermogenesis and adi-
synergistic modulation of brown adipocyte functions pose tissue remodelling148,150 (Fig. 5a). Adrenergic recep-
by sympathetic adrenergic and purinergic pathways has tors activate the classic cAMP signalling pathway that
been proposed143. Moreover, it has been revealed that activates protein kinase A (PKA), which activates lipo­
ATP is a necessary sympathetic co-​transmitter for the lysis of triacylglycerol within adipocyte lipid droplets in
high efficacy and specificity of noradrenaline-​induced WAT and BAT8,148. This pathway also induces the brown-
thermogenesis in brown adipocytes144. ing of WAT through the appearance of beige adipocytes
In response to sympathetic neurotransmitters, adipo- and the BAT thermogenic gene programme, enhancing
cytes and other adipose-​resident cells produce bioactive the expression of mitochondrial and oxidative genes,
factors, which modulate the functions of adipocytes, including the mitochondrial UCP1 (refs8,148,151).
including the cellular responses to the neurotransmitters The β-​adrenergic receptors (β1, β2 and β3 (Box 1))
themselves96,145–147 In this regard, the neurotransmitters are necessary mediators of noradrenaline’s actions on
secreted by adipose sympathetic nerve endings act in adipose tissue, as mouse models lacking these three
conjunction with other inputs originating from adipose-​ receptor subtypes have dysregulation of energy metabo­
resident cells and fine-​tune the adipocyte response to the lism and are prone to obesity152,153. Sustained release
neurotransmitters96,145–147 (Fig. 5). of noradrenaline and consequent receptor activation

www.nature.com/nrendo
Reviews

also exerts profound morphological changes in adi- functional role of these receptor subtypes on adipose
pose cellularity and tissue remodelling150. Among these and systemic metabolism needs further rigorous inves-
changes is the enhanced blood flow in BAT during tigation, applying both pharmacological and genetic
cold acclimation. The secretion of local vasodilating approaches to determine their full functional relevance.
factors, such as nitric oxide (NO), has been proposed
to increase blood flow in BAT during cold-​induced Adipose signals to sympathetic nerves
thermogenesis154,155. The sympathetic stimulus seems to Neurotrophic factors. It seems probable that adipose-​
participate in this process, as noradrenaline enhances resident cells affected by SNS secretions signal back
regional blood flow, along with increasing the expres- to local sympathetic nerve fibres within coordinated
sion and activity of the NO-​biosynthetic enzyme feedback responses167 (Fig.  5b). Accordingly, several
(NOS) in BAT156,157. Altogether, these dramatic changes WAT-​derived and BAT-​derived secreted factors that are
mediate the adaptive responses of adipose tissue to regulated by stimulation with noradrenaline have been
chronic exposure to low temperature or to metabolic reported to act on efferent nerves. Among these factors
stress, such as caloric restriction158. are the neurotrophic factors NRG4 (refs59–61), neuronal
growth regulator 1 (NEGR1)168, NGF63 and BDNF62.
Neuropeptide Y. Although NPY is largely produced Chronic β-​adrenergic receptor activation by noradren-
by CNS neurons, this neuropeptide is also synthesized by aline induces the formation of beige adipocytes in WAT,
peripheral sympathetic nerve fibres, acting as a neuro- which also produce some of these neuropeptides7. As
transmitter that affects adipose-​resident cells159 (Fig. 5a). these factors act as potent neuronal regulators, it is
NPY receptor subtypes 1 and 2 mediate the actions plausible that they enhance sympathetic innervation
of NPY and belong to the seven-​t ransmembrane in adipose tissue during cold-​induced thermogenesis
Gi/o protein-​coupled receptor family that inhibits ade- and browning. This positive loop of beige adipocytes
nylyl cyclase and thus the cAMP–PKA pathway, acting promoting their own formation through enhanced
antagonistically to adrenergic signalling159. Activation of sympathetic nerve density might be critical for adipose
the NPY receptor suppresses lipolysis in adipocytes160 thermogenesis. In support of this possibility, inhibiting
while also activating the extracellular-​signal-regulated NGF and its receptor TRKA suppresses browning of
kinase (ERK) pathway and adipogenesis161. Similar to adipose tissue63. This study suggests that NGF mediates
noradrenaline, NPY is located in cytosolic granules in sympathetic plasticity within adipose tissue through
the terminals of sympathetic neurons. In mice, its release enhanced innervation during cold exposure63. Such
and actions can be triggered by stressful situations, such neuronal plasticity resembles the active remodelling
as exposure to cold or overfeeding162. of adipose tissue depots during times of stress. As the
Importantly, signals emanating from NPY receptors TRKA protein is expressed in other tissues, it would be
promote adipose angiogenesis, along with differentia- important to selectively inactivate NGF–TRKA signal-
tion of new adipocytes162. In addition, obese mice were ling in sympathetic nerve endings in adipose tissue to
found to have elevated circulating levels of NPY and confirm the requirement for this pathway in adipose
increased expression of its receptors in WAT, which tissue remodelling during cold exposure.
suggests that this neuropeptide has a role in adipose tis-
sue accretion during obesity162. Indeed, this notion was Fatty acids and bioactive lipid metabolites. Although
further supported by adipose-​targeted knockdown and the effects of fatty acids and bioactive lipids on adipose
pharmacological inhibition of the NPY2 receptor, which sensory nerve fibres have been investigated, mecha-
resulted in reduced obesity and metabolic abnormali- nistic information on how these molecules affect adi-
ties in mice162. Altogether, it seems that NPY produced pose sympathetic nerves is limited (Fig. 5b). Lipolysis
by adipose sympathetic nerves functions in a system of of triacylglycerol and de novo biosynthesis of fatty
feedback responses that oppose the actions of noradren- acids in adipocytes might generate fatty acids and lipid
aline. Further studies to identify the signals that triggermetabolites that could act on adipose efferent fibres169.
the sympathetic production, release and actions of NPY Indeed, in vivo studies have shown that chronic infu-
are needed to clarify the exact role of NPY in regulating sion of fatty acids reduces peripheral sympathetic
systemic metabolism and the metabolic syndrome. activity in rodents170. However, this effect might be
through CNS regulation by fatty acids171, and whether
ATP. Similar to noradrenaline and NPY, ATP is a co-​ fatty acids also control peripheral sympathetic nerves
transmitter released by sympathetic nerve endings163 directly in a physiologically relevant manner remains
(Fig. 5a). When secreted from neurons, ATP exerts its to be determined.
effects via the ionotropic purinergic receptor sub-
types (P2XRs) and metabotropic purinergic receptor Endocannabinoids. Studies have demonstrated that
subtypes (P2YRs)164. Evidence suggests that signalling adipocytes produce substantial amounts of endocan-
through these purinergic receptors affects several differ­ nabinoids172–174 (Fig. 5b). Acting through their recep-
ent processes in adipose tissue, including lipid meta­ tors (CB1 and CB2), these bioactive lipids stimulate
bolism (enhanced lipogenesis and lipolysis), increased energy intake and inhibit energy expenditure, playing
thermogenesis, inflammation and endocrine func- an important role in energy balance175. Importantly,
tions164,165. For example, P2RX5 expression is induced dysregulation of endocannabinoid production in adi-
in BAT and WAT upon chronic cold exposure and pose tissue and CB1 activation might contribute to
correlates with UCP1 expression166. Nonetheless, the metabolic dysfunction172,175–178. Thus, in patients with

Nature Reviews | Endocrinology


Reviews

obesity, high circulating levels of endocannabinoids are in the liver, with lower amounts produced by other tis-
positively correlated with increased visceral fat176,177, sues187. However, under conditions of metabolic stress
which suggests that increased levels of endocannabi- such as obesity, the adipose expression level of GDF15
noids might contribute to metabolic dysregulation dur- is strongly upregulated184, which might be owing to
ing obesity. Furthermore, adipose-​specific inactivation increased amounts of ATM in the tissue. Although the
of CB1 seems to increase adipose thermogenesis178, effects of GDF15 on energy metabolism are known to
attenuating obesity-​induced metabolic dysfunction in be mediated through suppression of food intake mech-
mouse models175. anisms in the brain188,189, peripheral actions of GDF15,
Although some evidence indicates a direct action including sympathetic neuronal regulation, might also
of endocannabinoids on adipocytes, other studies have occur. Consistent with this notion, sympathetic neurons
suggested an inhibitory effect of endocannabinoids on express the GDF15 receptor GFRAL189, and signalling
neural circuits that blunt adipose sympathetic activ- through this receptor induces a potent neurotropic effect
ity and thermogenesis179,180. Although these effects are both in vitro and in vivo190. Nonetheless, it remains to be
probably mediated through central mechanisms, some tested whether adipose-​derived GDF15 has a direct role
reports suggest that endocannabinoids might also act as a peripheral neuronal modulator.
directly on peripheral sympathetic nerves181,182. In the
perfused mesenteric vascular bed of the rat, activation of Adenosine. A study published in 2014 indicated that
the CB1 receptor by the endocannabinoid anandamide adenosine, through its A2A receptors, induces BAT
suppresses the release of noradrenaline and ATP from and WAT thermogenesis191. This metabolite is released
sympathetic nerve terminals181. Altogether, these obser- from adipocytes or from other adipose-​resident cells
vations suggest that adipose-​derived fatty acids and bio- (Fig. 5b) . Moreover, adenosine can also be produced
active lipids such as the endocannabinoids might act on from extracellular ATP by Treg lymphocytes and possi-
local sympathetic fibres, suppressing their activity. Thus, bly endothelial cells, which express the enzymes ecto-
the increased biosynthesis of endocannabinoids in adi- 5ʹ-nucleotidases CD33 and CD73, cell surface proteins
pose tissue that has been noted in obesity might inhibit that are necessary for extracellular adenosine genera-
adipose sympathetic outflow, contributing to metabolic tion from ATP185. Importantly, the presence of adeno-
dysfunction. Endocannabinoids produced in adipose sine receptors was also detected in peripheral efferent
tissue might also signal through afferent nerve fibres, fibres in the vasculature, and signalling via these recep-
which should be studied in more detail. tors seems to blunt the secretion of neurotransmitters
in sympathetic nerve endings192,193, which suggests that
Cytokines and GDF15. Adipocytes are not the only adenosine within adipose tissue might also be involved
adipose-​t issue-resident cells that secrete neuronal in a negative feedback mechanism.
modulators that might affect adipose sympathetic nerve
fibres (Fig. 5b). Adipose tissue macrophages (ATMs) Sympathetic dysregulation in obesity
produce a number of pro-​i nflammatory and anti-​ General SNS dysregulation in metabolic disease is well
inflammatory cytokines, such as TNF, IL-1β, IL-6 and established194. As sympathetic outflow into adipose
IL-10, which are all reported to modulate sympathetic tissue controls such important metabolic processes in
nerve activity183. When produced in peripheral tissues, adipocytes34,133,134, it is not surprising that obesity dis-
cytokines can enter the circulation and gain access to rupts catecholamine-​mediated adipocyte lipolysis195,196,
the brain, enhancing the sympathetic outflow via CNS mitochondrial biogenesis197 and adipose tissue remodel-
regulation183. However, in chronic inflammatory states ling198. Sympathetic signals emanating from the CNS to
(as occurs in adipose tissue during obesity), increased adipose tissue might be altered in obesity by the associ-
local levels of inflammatory cytokines might restrict ated hyperinsulinaemia199, as insulin receptor signalling
sympathetic nerve fibres from regions of inflamed adi- in the brain is known to suppress adipose sympathetic
pose tissue or even damage nerves, depending on the outflow in lean mice134. Thus, chronic hyperinsulin­aemia
severity of inflammation183. This sympathetic neuronal might cause insufficient sympathetic activity within adi-
response during a persistent inflammatory state, as pose tissue, attenuating catecholamine-mediated actions.
occurs in arthritis, has been investigated in depth and Consistent with this notion, eliminating the hyperin-
previously described183. However, evidence supporting sulinaemia normally observed in mice fed a high-fat
the idea that sympathetic nerve repulsion occurs in diet by genetic deletion of three insulin alleles repro-
inflamed adipose tissue remains speculative, and experi­ grammes WAT to express UCP1 and increase energy
ments to rigorously test this hypothesis are necessary. expenditure199,200. Thus, pathways that are enhanced by
Thus, although adipose sympathetic outflow produces catecholamine are promoted upon reduction in circu-
an anti-​inflammatory response (see subsequent sec- lating levels of insulin. Chronic hyperinsulinaemia in
tion), the persistent cytokine production and chronic obesity also inhibits β-​adrenergic receptor signalling
inflammation noted in obesity might disrupt adipose through marked downregulation of β3-adrenergic recep-
efferent nerve function. tors, as noted in adipocytes from obese mice198,201. This
Other reported neuronal modulatory factors potenti­ decreased adipocyte β3-adrenergic receptor expression
ally produced by adipose-resident cells are growth differ­ and signalling is reversed by treatment of mice with
entiation factor 15 (GDF15)184 and adenosine185. Although diazoxide to reduce insulin secretion from β-cells202.
initially identified as a cytokine secreted by macro­ Although the inhibitory effect of central insulin
phages186, GDF15 seems to be expressed predominantly action on sympathetic outflow into WAT has been

www.nature.com/nrendo
Reviews

a b
Adipose tissue 1 2
SLC6A2
SNS
Noradrenaline MAOA
SAMs
4
1
3 β2-AR
M2 VEGF
2 M1
T cell
Macrophage ASC ↓ β-AR–PKA pathway ↑ Angiogenesis
↑ Macrophage polarization ↑ Sympathetic innervation

3 4 ATP Treg cell


Endothelium β1-AR

CD39
NGF CD73 Adenosine
BDNF
Adipocyte NRG4

P2R A2A
Systemic metabolism ↑ β-AR–PKA pathway
↑ Thermogenesis Heat ↑ UCP1
↑ Sympathetic innervation? Beige ↑ Thermogenesis
adipocyte

Fig. 6 | integrating peripheral signals in adipose tissue. a | Much like the integration of adipose signals within the brain,
neuro–adipose signal integration also occurs within the periphery. Examples include modulation of the adipose tissue
microenvironment via interaction with immune cell populations, the endothelium and adipose stem cells (ASCs).
Responses of adipose tissue to sympathetic nervous system (SNS) cues enable the rapid adaptation and remodelling
that is required to maintain systemic metabolic homeostasis. b | Sympathetic nerve fibres engage in unique interactions
with adipose tissue cell populations. Noradrenaline release from sympathetic terminals leads to adipose macrophage
polarization (Box 1) from a pro-​inflammatory (M1) to an anti-​inflammatory , alternatively activated (M2) profile.
Sympathetic neuron-​associated macrophages (SAMs) localize around sympathetic synapses and take up secreted
noradrenaline through the solute carrier family 6 member 2 (SLC6A2) noradrenaline transporter. Monoamine oxidase A
(MAOA) catalyses the degradation of noradrenaline within the SAMs (part 1). Noradrenaline release from sympathetic
nerve endings stimulates the β2-adrenergic receptor (β2-AR) in the vasculature, leading to vascular endothelial growth
factor (VEGF) secretion from the endothelium. VEGF stimulates angiogenesis and neurite outgrowth, driving increased
irrigation and innervation of the adipose tissue (part 2). ASC β1-AR activation by sympathetic noradrenaline drives beige
adipocyte differentiation. These beige adipocytes have an enhanced thermogenic capacity relative to white adipocytes
and a brown-​like adipokine expression profile, which might include neurotrophic factors such as nerve growth factor
(NGF), brain-​derived neurotrophic factor (BDNF) and neuregulin 4 (NRG4). These factors can drive increased sympathetic
innervation and arborization (part 3). The sympathetic co-​transmitter ATP is cleaved by regulatory T (Treg) cells into
adenosine via CD73-mediated and CD39-mediated degradation to create the anti-​inflammatory purinergic halo
surrounding the Treg cells. The adenosine interacts with the adenosine A2A (A2A) receptor to drive beige adipocyte
thermogenesis through mitochondrial UCP1 upregulation. Sympathetic-​derived ATP can also directly interact with the
purinergic P2 receptor family (P2R) to drive beige adipocyte thermogenesis (part 4). PKA , protein kinase A.

previously demonstrated by several studies122,134,200, it improved insulin signalling, along with an improve-
is important to emphasize that in other studies insu- ment in systemic metabolism79. However, whether
lin signalling in the brain has been claimed to increase these metabolic enhancements were associated with
sympathetic drive into peripheral tissues, such as kid- elevated adipose sympathetic tone and/or activity was
ney and BAT203,204. Thus, insulin signalling in the brain not investigated.
seems to elicit distinct sympathetic effects, depending
on the tissue. As central insulin signalling is disrupted Integrating adipose signalling networks
during metabolic dysfunction, it would be important to The many types of resident cell within adipose tissue
examine whether perturbation of brain insulin signal- communicate with each other through complex signal-
ling affects the increased sympathetic drive into some ling networks, as exemplified in Fig. 6a. Sympathetic
peripheral tissues. neuro­transmitters control key functions in endothelial
Similar to adipose sensory innervation, local sym- cells and multiple types of immune cell in addition to
pathetic innervation might also be affected by the per- adipocytes, and each of these cell types communicate
sistent adipose inflammation observed in obesity145. with adipocytes and each other, adjusting their meta-
Interestingly, moderate reduction in hyperinsulinaemia bolic pathways and shaping the tissue microenviron-
in obese mice decreased adipose inflammation and ment. This adipose tissue complexity also includes

Nature Reviews | Endocrinology


Reviews

external signals that are integrated within the tissue and increased SAM content in the adipose tissue, which
then relayed to other tissues from the adipose tissue in suppresses sympathetic drive and elevates adipose tis-
the form of secreted factors that act through the circu- sue inflammation145,147,212. These findings indicate that
lation or on sensory nerve fibres. Thus, adipose depots perturbations in neuron–immune cell interactions are
act as both sensors and integrators of complex signal- contributors to metabolic disease.
ling networks, and adipose innervation is critical to
this function. Neuron–endothelial cell communication
The close association between adipose nerve fibres
Neuron–immune cell communication and the vasculature indicates interdependency between
A variety of adipose-​resident immune cells express these two structures that might be essential for their
adrenoreceptors that can be activated upon the release functional role in regulating local and systemic metab-
of noradrenaline by adipose sympathetic nerve endings. olism213. Indeed, as shown in Fig. 6b, part 2, noradren-
ATMs represent one such example of cells that have a aline secreted by sympathetic nerve endings activates
pivotal role in adipose inflammation and systemic β-​adrenergic receptors in the endothelium to induce
metabolism25,79. ATMs can be sorted into two major sub- VEGF production214. In turn, endothelium-​derived
types, denoted M1 and M2, which exert either primarily VEGF215,216 might promote angiogenesis through stim-
pro-​inflammatory (that is, M1) or anti-​inflammatory ulation of endothelial cells and increase sympathetic
(that is, M2) functions205,206. Although macrophages innervation through activation of VEGFR signalling in
probably form a continuum of states between these two sympathetic neurons217,218. This physiological response
extreme types, several studies have shown that in healthy, of the endothelium to a sympathetic stimulus might be
functional adipose tissue, the ratio of M1-defined to essential in situations in which increased vasculariza-
M2-defined macrophages is low but is shifted towards an tion and innervation are needed, such as adipose tissue
M1 profile in unhealthy adipose tissue, as in obesity207,208. expansion during overfeeding31 or in cold-​induced ther-
Strategies to shift this ratio towards an M2-dominant mogenesis219. Accordingly, inhibition of VEGFR signal-
population seem to improve adipose and whole-​body ling in adipose-​resident cells disrupts thermogenesis and
metabolism209,210. Interestingly, catecholamines exert a browning in WAT induced by cold temperatures220–222.
potent effect on macrophages through β2-adrenergic Conversely, overexpression of the VEGF peptide elic-
receptors that promote M1 macrophage differentiation its browning in WAT76,220,223,224, which is consistent with
into the M2 state211, which indicates that sympathetic sig- VEGF having a key role in promoting adipose pathways
nals favour an anti-​inflammatory state (Fig. 6b, part 1). that benefit systemic metabolism.
This concept is supported by a study showing that sym-
pathetic nerve activity maintains an anti-​inflammatory Neuron–adipocyte progenitor communication
state in adipose tissue by inhibiting TNF expression in Adipose-​resident perivascular cells (Fig. 6b, part 3) are a
macrophages through activation of the β2-adrenergic subset of progenitor cells that differentiate into beige adi-
receptor–PKA pathway146. Conversely, adipose sympa- pocytes through activation of their β1-adrenergic recep-
thetic denervation leads to a marked increase in tissue tors225. This mechanism seems to be distinct from the
inflammation and dysfunction146. well-​known β3-adrenergic receptor-​induced browning
Surprisingly, a newly identified macrophage sub- of WAT, as it relies on the β1-adrenergic receptor rather
type (M1-like) seems to participate in a mechanism than the β3-adrenergic receptor isotype225. Interestingly,
to blunt sympathetic signalling in adipose and other Adrb1-null mice are intolerant of cold exposure and
tissues (Fig. 6b, part 1). These macrophages display a cannot defend their body temperature, highlighting
close anatomical and functional association with local the importance of β1-adrenergic receptors for adipose
sympathetic nerve fibres and are therefore named sym- thermogenesis and euthermia226. Additionally, these
pathetic neuron-​associated macrophages (SAMs)145,147. perivascular-​derived beige adipocytes not only display
Moreover, these macrophages exhibit high levels of increased thermogenic activity but also might secrete
a noradrenaline transporter (solute carrier family neurotrophic factors to promote adipose sympathetic
6 member 2 (SLC6A2)) in their plasma membranes innervation (Fig. 6b, part 3). Thus, sympathetic stim-
and are active in taking up noradrenaline released by ulation and differentiation of perivascular progenitor
sympathetic neurons and degrading it using the mon- cells into beige adipocytes might strongly contribute
oamine oxidase A (MAOA) pathway145,147. Interestingly, to browning of WAT during a cold stimulus, especially
genetic deletion of Slc6a2 in macrophages inhibits in mice that have not been previously exposed to the
noradrenaline uptake by SAMs, enhancing sympa- cold139. The importance of this mechanism remains
thetic signalling to the adipose tissue and improving to be determined by inactivating β1-adrenergic recep-
energy homeostasis145. Thus, the above pathways reveal tors in perivascular cells and examining the possible
tight communication between neurons and immune consequences for energy metabolism.
cells in adipose tissue: on the one hand, sympathetic
nerve endings signal to reduce adipose inflamma- Dysregulation of peripheral signals
tion by shifting the population of macrophages to an Similar to how hypothalamic dysregulation negatively
M2 phenotype; on the other hand, SAMs decrease the affects systemic metabolism, a failure in peripheral sig-
availability of released catecholamine and, therefore, nal integration can occur in obesity and might lead to
the sympathetic signal itself. Interestingly, obesity and metabolic dysfunction and T2DM5,10. Dysregulation of
age-​related metabolic dysfunction are associated with a homeostatic mechanism might arise from and result

www.nature.com/nrendo
Reviews

in the overproduction or underproduction of potent controlling the extracellular concentrations of ATP and
signalling molecules. The overproduction of NPY in adenosine. Therefore, adipose Treg cells and endothelial
obesity is one such case of signalling molecule dysreg- cells can modulate the tissue inflammation state by con-
ulation162. As detailed in a previous section, this neuro- trolling the conversion of extracellular ATP into adeno-
peptide favours adipose tissue expansion and has been sine (Fig. 6b, part 4). Disruptions of adipose endothelial
implicated in obesity-​linked metabolic dysfunction. function and reduction in Treg cell content, as noted in
Strategies such as pharmacologic inhibition and genetic obesity, would diminish ATP hydrolysis, increasing the
deletion aimed at blocking signalling from one of the ATP:adenosine ratio in the extracellular compartment.
NPY receptor subtypes (NPY2) were successful in atten- Consequently, high levels of ATP would activate M1
uating various metabolic abnormalities during obesity macrophages and favour the chronic inflammatory state
in mice162. Moreover, NPY signalling in vasculature typically seen in adipose tissue in obesity.
increased adhesion of leukocytes to human endothelial Hence, the inability of the adipose-​resident Treg cells
cells227,228, and treatment with a global NPY receptor and endothelial cells to control the extracellular levels
antagonist effectively improved chronic inflammation of ATP due to disruption of these cells might affect
in mice227,229. Thus, it is conceivable that persistent acti- the function of adipose tissue and proper integration
vation of adipose-​resident cells by increased NPY levels of peripheral signals to control systemic metabolism.
promotes adipose tissue accretion, favours immune cell Consistent with this notion, global genetic deletion of
infiltration and exacerbates adipose inflammation. CD39 leads to a decreased ability of endothelial cells to
Imbalance in adipose tissue due to persistent signals control extracellular ATP levels in the liver, enhanced
emanating from chronically activated NPY receptors in inflammation and systemic insulin resistance 234.
obesity might contribute to the disruption of the func- It will be important to examine the role of the adipose
tion of adipose tissue, such as local signal integration. CD39 and CD73 as modulators of ATP and adenosine
However, a study has suggested that NPY derived from signalling in adipose tissue metabolism and possible
macrophages and haematopoietic cells initiates an anti-​ influences in whole-​body metabolism.
inflammatory response during the early stages of obesity In summary, the ability of adipose tissue to integrate
in mice230. Accordingly, deletion of NPY in these cells central and peripheral signals relies on the accurate reg-
promotes inflammation in adipose tissue230. The reason ulation of adipose–neuron crosstalk and maintenance of
for the discrepancy between this study and the previous tissue homeostasis. In obesity, adipose expansion trig-
results could be the differential effect of NPY at different gers tissue inflammation, favouring pro-​inflammatory
stages of adipose tissue expansion in obesity, the origin M1 macrophages and diminishing the local sympathetic
of the NPY (nerve versus immune or other cell types) or signalling (Fig. 6b, part 1). The reduction in noradren-
the differential signalling by receptor 1 versus receptor 2. aline levels will not only favour a pro-​inflammatory
Additional studies will be necessary to better understand state but also blunt VEGF-​m ediated angiogenesis
these complexities of NPY function in adipose tissue. and adipose innervation (Fig. 6b, part 2). Lower levels
ATP is a sympathetic co-​transmitter; dysregulation of noradrenaline might also reduce differentiation of
of its production and/or its degradation by adipose-​ perivascular-​derived beige adipocytes, which affects
resident cells might be implicated in obesity-​linked adi- adipose thermogenesis and potentially innervation
pose dysfunction and disruption of adipose signalling (Fig. 6b, part 3). Chronic adipose inflammation might
integration231. An increase in the extracellular levels of also reduce vascular function and the presence of anti-​
ATP is often associated with tissue inflammation and inflammatory Treg lymphocytes, resulting in extracellular
the metabolic abnormalities that occur in adipose tis- accumulation of ATP and persistent tissue inflammation
sue during obesity231,232. Consistent with this notion, (Fig. 6b, part 4). Fine-​tuned regulation of these processes
the adipose tissue from individuals with obesity who might be essential for adipose–peripheral integration
are metabolically unhealthy releases increased levels of and ultimately whole-​b ody metabolic homeostasis.
ATP compared with adipose tissue from lean and meta­ A failure to appropriately regulate such processes might
bolically healthy individuals231. Such an increase in the be linked to metabolic disorders in obesity and T2DM.
extracellular levels of ATP stimulates purinergic recep-
tor P2RX7 signalling in M1 macrophages to increase Conclusions
pro-​inflammatory cytokine production and adipose Findings showing dynamic crosstalk between adipose-​
inflammation233. Therefore, local mechanisms that con- resident cells and local nerve fibres have opened a fer-
trol the extracellular ATP levels are essential for tissue tile area of adipose biology for more detailed study. The
homeostasis. One such local mechanism that has a role revelation that lipolysis of fat stores, which is required
in regulating extracellular concentrations of ATP is the for survival in fasting, is stimulated by noradrenaline
cleavage of ATP into adenosine. released from efferent, sympathetic nerve fibres within
In contrast to the pro-​inflammatory role of extracel- adipose tissue rather than by circulating catecholamines
lular ATP, adenosine produced by ATP degradation is reinforces the importance of this topic133. Even the dra-
known to limit severe inflammation and tissue damage232. matic suppression of adipocyte lipolysis during feeding
The secreted ATP can be metabolized into adenosine by by insulin involves regulation of sympathetic tone in adi-
two extracellular ecto-5ʹ-nucleotidases — CD39 and pose tissue122,134. Similarly, it is plausible that afferent, sen-
CD73 (ref.232). These two enzymes, which are expressed sory nerve fibres might also modulate adipose-​resident
at the cell surface of Treg cells84,85 and in the adipose cells, providing an exciting area for further research
vasculature234 (Fig. 6b, part 4), might have a key role in to identify factors that mediate such putative effects.

Nature Reviews | Endocrinology


Reviews

Conversely, it is now appreciated that communication NPY and ATP that is associated with metabolic disease
moves in the other direction as well: adipocytes is also linked to chronic adipose dysfunction and the
and other adipose-​resident cell types can signal to failure of adipose tissue to regulate systemic metabolism.
nearby sensory nerve fibres that transmit information Obesity and pro-​inflammatory cytokines also reduce the
to the CNS and to sympathetic neurons that control biosynthesis of adipocyte-​derived neurotrophic factors,
SNS tone. Some factors secreted from adipocytes that such as NRG4 (refs64,235), which has also been shown to
act on sensory neurons have been identified, including be a beneficial circulating agent that controls glucose
leptin and fatty acids, but this area of investigation is still homeostasis and liver fat metabolism. Similarly, there is
at an early stage and offers considerable opportunities high interest in identifying what triggers hypothalamic
for discovery. Importantly, much of the data available inflammation and to what extent it causes metabolic
and cited in this Review relate to mouse studies, and it dysfunction in obesity. Bioactive lipids, glycosylated pro-
is critical for the field to test these findings and ideas in teins, pro-​inflammatory cytokines and perhaps hyper-
human tissues. insulinaemia are top candidates among the suspected
Another fertile area for investigation relates to the factors that might elicit hypothalamic inflammation. As
key role of the endothelium, adipose progenitor cells this research area is still fairly new, taking advantage of
and immune cells as both targets of neuronal signals new techniques and developing more refined methods
within adipose tissue and as generators of signals to will be critical to optimize progress in understanding
local nerve fibres. Noradrenaline and the neurotrans- communication between adipose-​resident cells and local
mitters CGRP and substance P are vasoactive factors and nerve fibres. Rather than relying on genetic deletion of
immune modulators that can regulate the production of neuronal receptors or ligands in all sensory or sympa-
vasoactive peptides and cytokines in the endothelium thetic nerve fibres, more localized perturbations and
and immune cells within adipose tissue when released reporters will be required. Optogenetic55,236 and chemical
by sympathetic and sensory nerve endings, respectively. genetic approaches237 (for example, designer receptors
Importantly, dysregulation of CGRP and substance P exclusively activated by designer drugs) will be valuable
might contribute to endothelial dysfunctions and per- approaches for future studies.
sistent adipose inflammation. This immune reaction Finally, the discovery that WAT metabolic activ-
resembles the neurogenic inflammation noted in other ity can modulate the function of distant BAT depots
tissues in type 1 diabetes mellitus and T2DM and, like- through sensory nerve signalling to the CNS to enhance
wise, might lead to adipose neuropathy (Box 1). However, sympathetic outflow represents an exciting future direc-
the extent to which adipose sensory nerve dysregulation tion for research in this field39,40,48,49. Coupled with exten-
and afferent nerve-​induced neurogenic inflammation sive data238 showing that the CNS can regulate metabolic
contribute to immune cell expansion in adipose tissue tissues, including liver, skeletal muscle and pancreatic
in obesity is unknown. Might targeting adipose neuro- islets, to control systemic glucose homeostasis, these
genic inflammation by blocking CGRP and substance P findings infer that adipose tissue might also signal to
receptors attenuate chronic adipose inflammation and these other tissues through the brain. Much more work
thus improve systemic metabolism in obesity? is required to test this idea and to define its full impor-
Adipose tissue innervation and the regulation of adi- tance, but hopefully this hypothesis will be enticing to
pose function by the CNS are probably important targets many scientists in the field.
for disruption by obesity and T2DM. Imbalance in the
levels of sympathetic neurotransmitters noradrenaline, Published online xx xx xxxx

1. Shaw, H. B. A. Contribution to the study of the resistance and type 2 diabetes. Nat. Rev. Mol. Cell 19. Dresner, A. et al. Effects of free fatty acids on glucose
morphology of adipose tissue. J. Anat. Physiol. 36, Biol. 9, 367–377 (2008). transport and IRS-1-associated phosphatidylinositol
1–13 (1901). 11. Nye, C., Kim, J., Kalhan, S. C. & Hanson, R. W. 3-kinase activity. J. Clin. Invest. 103, 253–259
2. Kershaw, E. E. & Flier, J. S. Adipose tissue as an Reassessing triglyceride synthesis in adipose tissue. (1999).
endocrine organ. J. Clin. Endocrinol. Metab. 89, Trends Endocrinol. Metab. 19, 356–361 (2008). 20. Petersen, K. F. & Shulman, G. I. Cellular mechanism of
2548–2556 (2004). 12. Teusink, B. et al. Contribution of fatty acids released insulin resistance in skeletal muscle. J. R. Soc. Med.
3. Ouchi, N., Parker, J. L., Lugus, J. J. & Walsh, K. from lipolysis of plasma triglycerides to total plasma 95 (Suppl. 42), 8–13 (2002).
Adipokines in inflammation and metabolic disease. fatty acid flux and tissue-​specific fatty acid uptake. 21. DeFronzo, R. A., Bonadonna, R. C. & Ferrannini, E.
Nat. Rev. Immunol. 11, 85–97 (2011). Diabetes 52, 614–620 (2003). Pathogenesis of NIDDM. A balanced overview.
4. Villarroya, F., Cereijo, R., Villarroya, J. & Giralt, M. 13. Kersten, S. Physiological regulation of lipoprotein Diabetes Care 15, 318–368 (1992).
Brown adipose tissue as a secretory organ. Nat. Rev. lipase. Biochim. Biophys. Acta 1841, 919–933 (2014). 22. Walther, T. C., Chung, J. & Farese, R. V. Jr. Lipid
Endocrinol. 13, 26–35 (2017). 14. Czech, M. P., Tencerova, M., Pedersen, D. J. droplet biogenesis. Annu. Rev. Cell Dev. Biol. 33,
5. Stern, J. H., Rutkowski, J. M. & Scherer, P. E. & Aouadi, M. Insulin signalling mechanisms for 491–510 (2017).
Adiponectin, leptin, and fatty acids in the maintenance triacylglycerol storage. Diabetologia 56, 949–964 23. Cinti, S. et al. Adipocyte death defines macrophage
of metabolic homeostasis through adipose tissue (2013). localization and function in adipose tissue of obese
crosstalk. Cell Metab. 23, 770–784 (2016). 15. Unger, R. H., Clark, G. O., Scherer, P. E. & Orci, L. mice and humans. J. Lipid Res. 46, 2347–2355
6. Shin, J. et al. SDF-1 is an autocrine insulin-​ Lipid homeostasis, lipotoxicity and the metabolic (2005).
desensitizing factor in adipocytes. Diabetes 67, syndrome. Biochim. Biophys. Acta 1801, 209–214 24. Kanda, H. et al. MCP-1 contributes to macrophage
1068–1078 (2018). (2010). infiltration into adipose tissue, insulin resistance,
7. Villarroya, F., Gavalda-​Navarro, A., Peyrou, M., 16. Perry, R. J. et al. Hepatic acetyl CoA links adipose and hepatic steatosis in obesity. J. Clin. Invest. 116,
Villarroya, J. & Giralt, M. The lives and times of brown tissue inflammation to hepatic insulin resistance and 1494–1505 (2006).
adipokines. Trends Endocrinol. Metab. 28, 855–867 type 2 diabetes. Cell 160, 745–758 (2015). 25. Amano, S. U. et al. Local proliferation of macrophages
(2017). 17. Titchenell, P. M. et al. Direct hepatocyte insulin contributes to obesity-​associated adipose tissue
8. Cannon, B. & Nedergaard, J. Brown adipose tissue: signaling is required for lipogenesis but is dispensable inflammation. Cell Metab. 19, 162–171 (2014).
function and physiological significance. Physiol. Rev. for the suppression of glucose production. Cell Metab. 26. Weisberg, S. P. et al. Obesity is associated with
84, 277–359 (2004). 23, 1154–1166 (2016). macrophage accumulation in adipose tissue. J. Clin.
9. Rosen, E. D. & Spiegelman, B. M. What we talk about 18. Clerk, L. H., Rattigan, S. & Clark, M. G. Lipid infusion Invest. 112, 1796–1808 (2003).
when we talk about fat. Cell 156, 20–44 (2014). impairs physiologic insulin-​mediated capillary 27. Xu, H. et al. Chronic inflammation in fat plays a crucial
10. Guilherme, A., Virbasius, J. V., Puri, V. & Czech, M. P. recruitment and muscle glucose uptake in vivo. role in the development of obesity-​related insulin
Adipocyte dysfunctions linking obesity to insulin Diabetes 51, 1138–1145 (2002). resistance. J. Clin. Invest. 112, 1821–1830 (2003).

www.nature.com/nrendo
Reviews

28. Gustafson, B. & Smith, U. Cytokines promote Wnt 53. Murphy, K. T. et al. Leptin-​sensitive sensory nerves 78. Dhondt, J. et al. Neuronal FLT1 receptor and its
signaling and inflammation and impair the normal innervate white fat. Am. J. Physiol. Endocrinol. Metab. selective ligand VEGF-​B protect against retrograde
differentiation and lipid accumulation in 3T3-L1 304, E1338–E1347 (2013). degeneration of sensory neurons. FASEB J. 25,
preadipocytes. J. Biol. Chem. 281, 9507–9516 54. de Lartigue, G., Ronveaux, C. C. & Raybould, H. E. 1461–1473 (2011).
(2006). Deletion of leptin signaling in vagal afferent neurons 79. Pedersen, D. J. et al. A major role of insulin in
29. Grant, R. W. & Stephens, J. M. Fat in flames: influence results in hyperphagia and obesity. Mol. Metab. 3, promoting obesity-​associated adipose tissue
of cytokines and pattern recognition receptors on 595–607 (2014). inflammation. Mol. Metab. 4, 507–518 (2015).
adipocyte lipolysis. Am. J. Physiol. Endocrinol. Metab. 55. Zeng, W. et al. Sympathetic neuro-​adipose connections 80. Hardy, O. T. et al. Body mass index-​independent
309, E205–E213 (2015). mediate leptin-​driven lipolysis. Cell 163, 84–94 inflammation in omental adipose tissue associated
30. Aouadi, M. et al. Gene silencing in adipose tissue (2015). with insulin resistance in morbid obesity. Surg. Obes.
macrophages regulates whole-​body metabolism in 56. Shiuchi, T. et al. Induction of glucose uptake in skeletal Relat. Dis. 7, 60–67 (2011).
obese mice. Proc. Natl Acad. Sci. USA 110, muscle by central leptin is mediated by muscle beta2- 81. Zanos, T. P. et al. Identification of cytokine-​specific
8278–8283 (2013). adrenergic receptor but not by AMPK. Sci. Rep. 7, sensory neural signals by decoding murine vagus
31. Sung, H. K. et al. Adipose vascular endothelial growth 15141 (2017). nerve activity. Proc. Natl Acad. Sci. USA 115,
factor regulates metabolic homeostasis through 57. Waxman, S. G. From Neuroscience to Neurology: E4843–E4852 (2018).
angiogenesis. Cell Metab. 17, 61–72 (2013). Neuroscience, Molecular Medicine, and the Therapeutic 82. Osamura, N. et al. Induction of interleukin-6 in
32. Halaas, J. L. et al. Weight-​reducing effects of the Transformation of Neurology (Elsevier Academic Press, dorsal root ganglion neurons after gradual elongation
plasma protein encoded by the obese gene. Science 2005). of rat sciatic nerve. Exp. Neurol. 195, 61–70
269, 543–546 (1995). 58. Korsching, S. The neurotrophic factor concept: (2005).
33. Bartness, T. J., Kay Song, C., Shi, H., Bowers, R. R. a reexamination. J. Neurosci. 13, 2739–2748 83. Miller, R. J., Jung, H., Bhangoo, S. K. & White, F. A.
& Foster, M. T. Brain-​adipose tissue cross talk. (1993). Cytokine and chemokine regulation of sensory neuron
Proc. Nutr. Soc. 64, 53–64 (2005). 59. Christian, M. Transcriptional fingerprinting of function. Handb. Exp. Pharmacol. 194, 417–449
34. Bartness, T. & Kay Song, C. Innervation of brown “browning” white fat identifies NRG4 as a novel (2009).
adipose tissue and its role in thermogenesis. adipokine. Adipocyte 4, 50–54 (2015). 84. Nunez Ruiz, A. et al. Diminished levels of regulatory
Can. J. Diabetes 29, 420–428 (2005). 60. Rosell, M. et al. Brown and white adipose tissues: T cell subsets (CD8+Foxp3, CD4+Foxp3 and
35. Bartness, T. J. & Ryu, V. Neural control of white, intrinsic differences in gene expression and response CD4+CD39+Foxp3) but increased Foxp3 expression
beige and brown adipocytes. Int. J. Obes. Suppl. 5, to cold exposure in mice. Am. J. Physiol. Endocrinol. in adipose tissue from overweight subjects. Nutrition
S35–S39 (2015). Metab. 306, E945–E964 (2014). 32, 943–954 (2016).
36. Caron, A., Lee, S., Elmquist, J. K. & Gautron, L. Leptin 61. Pellegrinelli, V. et al. Adipocyte-​secreted BMP8b 85. Cortez-​Espinosa, N. et al. CD39 expression on Treg
and brain-​adipose crosstalks. Nat. Rev. Neurosci. 19, mediates adrenergic-​induced remodeling of the neuro-​ and Th17 cells is associated with metabolic factors in
153–165 (2018). vascular network in adipose tissue. Nat. Commun. 9, patients with type 2 diabetes. Hum. Immunol. 76,
37. Friedman, J. 20 years of leptin: leptin at 20: 4974 (2018). 622–630 (2015).
an overview. J. Endocrinol. 223, T1–T8 (2014). 62. Nakagomi, A. et al. Role of the central nervous system 86. Herbert, M. K. & Holzer, P. Neurogenic inflammation. I.
38. Zhang, Y. et al. Positional cloning of the mouse obese and adipose tissue BDNF/TrkB axes in metabolic Basic mechanisms, physiology and pharmacology
gene and its human homologue. Nature 372, regulation. NPJ Aging Mech. Dis. 1, 15009 (2015). [German]. Anasthesiol. Intensivmed. Notfallmed.
425–432 (1994). 63. Cao, Y., Wang, H. & Zeng, W. Whole-​tissue 3D imaging Schmerzther. 37, 314–325 (2002).
39. Ryu, V., Garretson, J. T., Liu, Y., Vaughan, C. H. reveals intra-​adipose sympathetic plasticity regulated 87. Russell, F. A., King, R., Smillie, S. J., Kodji, X.
& Bartness, T. J. Brown adipose tissue has by NGF-​TrkA signal in cold-​induced beiging. Protein & Brain, S. D. Calcitonin gene-​related peptide:
sympathetic-​sensory feedback circuits. J. Neurosci. Cell 9, 527–539 (2018). physiology and pathophysiology. Physiol. Rev. 94,
35, 2181–2190 (2015). 64. Wang, G. X. et al. The brown fat-​enriched secreted 1099–1142 (2014).
40. Garretson, J. T. et al. Lipolysis sensation by white factor Nrg4 preserves metabolic homeostasis through 88. Brain, S. D. & Grant, A. D. Vascular actions of
fat afferent nerves triggers brown fat thermogenesis. attenuation of hepatic lipogenesis. Nat. Med. 20, calcitonin gene-​related peptide and adrenomedullin.
Mol. Metab. 5, 626–634 (2016). 1436–1443 (2014). Physiol. Rev. 84, 903–934 (2004).
41. Harris, R. B. S. Denervation as a tool for testing 65. Min, S. Y. et al. Human ‘brite/beige’ adipocytes 89. Sung, C. P. et al. CGRP stimulates the adhesion of
sympathetic control of white adipose tissue. Physiol. develop from capillary networks, and their leukocytes to vascular endothelial cells. Peptides 13,
Behav. 190, 3–10 (2018). implantation improves metabolic homeostasis in mice. 429–434 (1992).
42. Fishman, R. B. & Dark, J. Sensory innervation of white Nat. Med. 22, 312–318 (2016). 90. Liu, T. et al. Endogenous calcitonin gene-​related
adipose tissue. Am. J. Physiol. 253, R942–R944 66. Pearson, R. J. Jr & Carroll, S. L. ErbB transmembrane peptide regulates lipid metabolism and energy
(1987). tyrosine kinase receptors are expressed by sensory homeostasis in male mice. Endocrinology 158,
43. Shi, H., Song, C. K., Giordano, A., Cinti, S. and motor neurons projecting into sciatic nerve. 1194–1206 (2017).
& Bartness, T. J. Sensory or sympathetic white adipose J. Histochem. Cytochem. 52, 1299–1311 (2004). 91. Walker, C. S. et al. Mice lacking the neuropeptide
tissue denervation differentially affects depot growth 67. Vetter, I., Pujic, Z. & Goodhill, G. J. The response of alpha-​calcitonin gene-​related peptide are protected
and cellularity. Am. J. Physiol. Regul. Integr. Comp. dorsal root ganglion axons to nerve growth factor against diet-​induced obesity. Endocrinology 151,
Physiol. 288, R1028–R1037 (2005). gradients depends on spinal level. J. Neurotrauma 27, 4257–4269 (2010).
44. De Matteis, R., Ricquier, D. & Cinti, S. TH-, NPY-, SP-, 1379–1386 (2010). 92. Aveseh, M., Koushkie-​Jahromi, M., Nemati, J.
and CGRP-​immunoreactive nerves in interscapular 68. Moy, J. K., Khoutorsky, A., Asiedu, M. N., Dussor, G. & Esmaeili-​Mahani, S. Serum calcitonin gene-​related
brown adipose tissue of adult rats acclimated at & Price, T. J. eIF4E phosphorylation influences Bdnf peptide facilitates adipose tissue lipolysis during
different temperatures: an immunohistochemical mRNA translation in mouse dorsal root ganglion exercise via PIPLC/IP3 pathways. Endocrine 61,
study. J. Neurocytol. 27, 877–886 (1998). neurons. Front. Cell. Neurosci. 12, 29 (2018). 462–472 (2018).
45. Song, C. K., Schwartz, G. J. & Bartness, T. J. 69. Cannon, B. et al. ‘Neuropeptide tyrosine’ (NPY) is 93. Johnson, M. B., Young, A. D. & Marriott, I. The
Anterograde transneuronal viral tract tracing reveals co-​stored with noradrenaline in vascular but not in therapeutic potential of targeting substance P/NK-1R
central sensory circuits from white adipose tissue. parenchymal sympathetic nerves of brown adipose interactions in inflammatory CNS disorders. Front.
Am. J. Physiol. Regul. Integr. Comp. Physiol. 296, tissue. Exp. Cell Res. 164, 546–550 (1986). Cell. Neurosci. 10, 296 (2016).
R501–R511 (2009). 70. Slavin, B. G. & Ballard, K. W. Morphological studies 94. Garcia-​Recio, S. & Gascon, P. Biological and
46. Vaughan, C. H. & Bartness, T. J. Anterograde on the adrenergic innervation of white adipose tissue. pharmacological aspects of the NK1-receptor. Biomed.
transneuronal viral tract tracing reveals central sensory Anat. Rec. 191, 377–389 (1978). Res. Int. 2015, 495704 (2015).
circuits from brown fat and sensory denervation alters 71. Bartness, T. J. & Bamshad, M. Innervation of 95. O’Connor, T. M. et al. The role of substance P in
its thermogenic responses. Am. J. Physiol. Regul. mammalian white adipose tissue: implications for inflammatory disease. J. Cell. Physiol. 201, 167–180
Integr. Comp. Physiol. 302, R1049–R1058 (2012). the regulation of total body fat. Am. J. Physiol. 275, (2004).
47. Ryu, V. & Bartness, T. J. Short and long sympathetic-​ R1399–R1411 (1998). 96. Reilly, S. M. & Saltiel, A. R. Adapting to obesity with
sensory feedback loops in white fat. Am. J. Physiol. 72. Carmeliet, P. & Tessier-​Lavigne, M. Common adipose tissue inflammation. Nat. Rev. Endocrinol. 13,
Regul. Integr. Comp. Physiol. 306, R886–R900 mechanisms of nerve and blood vessel wiring. Nature 633–643 (2017).
(2014). 436, 193–200 (2005). 97. Ellis, A. & Bennett, D. L. Neuroinflammation and the
48. Nguyen, N. L. T., Xue, B. & Bartness, T. J. Sensory 73. Shvartsman, D. et al. Sustained delivery of VEGF generation of neuropathic pain. Br. J. Anaesth. 111,
denervation of inguinal white fat modifies sympathetic maintains innervation and promotes reperfusion in 26–37 (2013).
outflow to white and brown fat in Siberian hamsters. ischemic skeletal muscles via NGF/GDNF signaling. 98. Yadav, R. L. et al. Somatic neural alterations in non-​
Physiol. Behav. 190, 28–33 (2018). Mol. Ther. 22, 1243–1253 (2014). diabetic obesity: a cross-​sectional study. BMC Obes.
49. Guilherme, A. et al. Neuronal modulation of brown 74. Mackenzie, F. & Ruhrberg, C. Diverse roles for 3, 50 (2016).
adipose activity through perturbation of white VEGF-​A in the nervous system. Development 139, 99. Richardson, J. D. & Vasko, M. R. Cellular mechanisms
adipocyte lipogenesis. Mol. Metab. 16, 116–125 1371–1380 (2012). of neurogenic inflammation. J. Pharmacol. Exp. Ther.
(2018). 75. Guaiquil, V. H. et al. VEGF-​B selectively regenerates 302, 839–845 (2002).
50. Pereira, M. M. et al. A brain-​sparing diphtheria toxin injured peripheral neurons and restores sensory and 100. Xanthos, D. N. & Sandkuhler, J. Neurogenic
for chemical genetic ablation of peripheral cell lineages. trophic functions. Proc. Natl Acad. Sci. USA 111, neuroinflammation: inflammatory CNS reactions in
Nat. Commun. 8, 14967 (2017). 17272–17277 (2014). response to neuronal activity. Nat. Rev. Neurosci. 15,
51. Niijima, A. Afferent signals from leptin sensors in the 76. Park, J. et al. VEGF-​A-expressing adipose tissue 43–53 (2014).
white adipose tissue of the epididymis, and their reflex shows rapid beiging and enhanced survival after 101. Schaper, N. C., Huijberts, M. & Pickwell, K.
effect in the rat. J. Auton. Nerv. Syst. 73, 19–25 transplantation and confers IL-4-independent Neurovascular control and neurogenic inflammation in
(1998). metabolic improvements. Diabetes 66, 1479–1490 diabetes. Diabetes Metab. Res. Rev. 24, S40–S44
52. Niijima, A. Reflex effects from leptin sensors in the (2017). (2008).
white adipose tissue of the epididymis to the efferent 77. Sun, K. et al. Dichotomous effects of VEGF-​A on 102. Farkas, G. J. & Gater, D. R. Neurogenic obesity and
activity of the sympathetic and vagus nerve in the rat. adipose tissue dysfunction. Proc. Natl Acad. Sci. USA systemic inflammation following spinal cord injury:
Neurosci. Lett. 262, 125–128 (1999). 109, 5874–5879 (2012). a review. J. Spinal Cord Med. 41, 378–387 (2017).

Nature Reviews | Endocrinology


Reviews

103. Morrison, S. F., Madden, C. J. & Tupone, D. 129. Baufeld, C., Osterloh, A., Prokop, S., Miller, K. R. 154. Nisoli, E., Tonello, C., Briscini, L. & Carruba, M. O.
Central neural regulation of brown adipose tissue & Heppner, F. L. High-​fat diet-​induced brain region-​ Inducible nitric oxide synthase in rat brown adipocytes:
thermogenesis and energy expenditure. Cell Metab. specific phenotypic spectrum of CNS resident implications for blood flow to brown adipose tissue.
19, 741–756 (2014). microglia. Acta Neuropathol. 132, 361–375 (2016). Endocrinology 138, 676–682 (1997).
104. Morrison, S. F. Central neural control of 130. Schur, E. A. et al. Radiologic evidence that 155. Petrovic, V. et al. NO modulates the molecular basis of
thermoregulation and brown adipose tissue. Auton. hypothalamic gliosis is associated with obesity and rat interscapular brown adipose tissue thermogenesis.
Neurosci. 196, 14–24 (2016). insulin resistance in humans. Obesity 23, 2142–2148 Comp. Biochem. Physiol. C 152, 147–159 (2010).
105. Williams, K. W. & Elmquist, J. K. From neuroanatomy (2015). 156. Takahashi, H. et al. Beta-3 adrenergic agonist,
to behavior: central integration of peripheral signals 131. Valdearcos, M. et al. Microglial inflammatory signaling BRL-26830A, and alpha/beta blocker, arotinolol,
regulating feeding behavior. Nat. Neurosci. 15, orchestrates the hypothalamic immune response to markedly increase regional blood flow in the brown
1350–1355 (2012). dietary excess and mediates obesity susceptibility. adipose tissue in anesthetized rats. Jpn Circ. J. 56,
106. Konner, A. C., Klockener, T. & Bruning, J. C. Control Cell Metab. 26, 185–197 (2017). 936–942 (1992).
of energy homeostasis by insulin and leptin: targeting 132. Giordano, A. et al. White adipose tissue lacks significant 157. Giordano, A. et al. Evidence for a functional nitric
the arcuate nucleus and beyond. Physiol. Behav. 97, vagal innervation and immunohistochemical evidence oxide synthase system in brown adipocyte nucleus.
632–638 (2009). of parasympathetic innervation. Am. J. Physiol. Regul. FEBS Lett. 514, 135–140 (2002).
107. Timper, K. & Bruning, J. C. Hypothalamic circuits Integr. Comp. Physiol. 291, R1243–R1255 (2006). 158. Sipe, L. M. et al. Differential sympathetic outflow to
regulating appetite and energy homeostasis: pathways 133. Bartness, T. J., Liu, Y., Shrestha, Y. B. & Ryu, V. Neural adipose depots is required for visceral fat loss in
to obesity. Dis. Model. Mech. 10, 679–689 (2017). innervation of white adipose tissue and the control response to calorie restriction. Nutr. Diabetes 7,
108. Hollenberg, A. N. The role of the thyrotropin-​releasing of lipolysis. Front. Neuroendocrinol. 35, 473–493 e260 (2017).
hormone (TRH) neuron as a metabolic sensor. Thyroid (2014). 159. Zhang, W., Cline, M. A. & Gilbert, E. R. Hypothalamus-​
18, 131–139 (2008). 134. Scherer, T. et al. Brain insulin controls adipose tissue adipose tissue crosstalk: neuropeptide Y and the
109. Ryu, V., Watts, A. G., Xue, B. & Bartness, T. J. lipolysis and lipogenesis. Cell Metab. 13, 183–194 regulation of energy metabolism. Nutr. Metab. 11, 27
Bidirectional crosstalk between the sensory and (2011). (2014).
sympathetic motor systems innervating brown and 135. Vitali, A. et al. The adipose organ of obesity-​prone 160. Turtzo, L. C., Marx, R. & Lane, M. D. Cross-​talk between
white adipose tissue in male Siberian hamsters. C57BL/6J mice is composed of mixed white and sympathetic neurons and adipocytes in coculture. Proc.
Am. J. Physiol. Regul. Integr. Comp. Physiol. 312, brown adipocytes. J. Lipid Res. 53, 619–629 (2012). Natl Acad. Sci. USA 98, 12385–12390 (2001).
R324–R337 (2017). 136. Jiang, H., Ding, X., Cao, Y., Wang, H. & Zeng, W. 161. Yang, K., Guan, H., Arany, E., Hill, D. J. & Cao, X.
110. Nguyen, N. L., Randall, J., Banfield, B. W. Dense intra-​adipose sympathetic arborizations are Neuropeptide Y is produced in visceral adipose tissue
& Bartness, T. J. Central sympathetic innervations essential for cold-​induced beiging of mouse white and promotes proliferation of adipocyte precursor
to visceral and subcutaneous white adipose tissue. adipose tissue. Cell Metab. 26, 686–692 (2017). cells via the Y1 receptor. FASEB J. 22, 2452–2464
Am. J. Physiol. Regul. Integr. Comp. Physiol. 306, 137. Chi, J. et al. Three-​dimensional adipose tissue imaging (2008).
R375–R386 (2014). reveals regional variation in beige fat biogenesis and 162. Kuo, L. E. et al. Neuropeptide Y acts directly in the
111. Bamshad, M., Aoki, V. T., Adkison, M. G., Warren, W. S. PRDM16-dependent sympathetic neurite density. periphery on fat tissue and mediates stress-​induced
& Bartness, T. J. Central nervous system origins Cell Metab. 27, 226–236 (2018). obesity and metabolic syndrome. Nat. Med. 13,
of the sympathetic nervous system outflow to white 138. Schulz, T. J. et al. Brown-​fat paucity due to impaired 803–811 (2007).
adipose tissue. Am. J. Physiol. 275, R291–R299 BMP signalling induces compensatory browning of 163. Burnstock, G. Purinergic cotransmission. Exp. Physiol.
(1998). white fat. Nature 495, 379–383 (2013). 94, 20–24 (2009).
112. Morton, G. J. & Schwartz, M. W. Leptin and the 139. Zhu, Y. et al. Connexin 43 mediates white adipose 164. Burnstock, G. & Gentile, D. The involvement of
central nervous system control of glucose metabolism. tissue beiging by facilitating the propagation of purinergic signalling in obesity. Purinergic Signal. 14,
Physiol. Rev. 91, 389–411 (2011). sympathetic neuronal signals. Cell Metab. 24, 97–108 (2018).
113. Contreras, C., Nogueiras, R., Dieguez, C., Rahmouni, K. 420–433 (2016). 165. Tozzi, M. & Novak, I. Purinergic receptors in adipose
& Lopez, M. Traveling from the hypothalamus to the 140. Burnstock, G. & Sneddon, P. Evidence for ATP and tissue as potential targets in metabolic disorders.
adipose tissue: the thermogenic pathway. Redox Biol. noradrenaline as cotransmitters in sympathetic Front. Pharmacol. 8, 878 (2017).
12, 854–863 (2017). nerves. Clin. Sci. 68, 89s–92s (1985). 166. Ussar, S. et al. ASC-1, PAT2, and P2RX5 are cell
114. Myers, M. G. Jr & Olson, D. P. Central nervous system 141. Pablo Huidobro-​Toro, J. & Veronica Donoso, M. surface markers for white, beige, and brown
control of metabolism. Nature 491, 357–363 (2012). Sympathetic co-​transmission: the coordinated action adipocytes. Sci. Transl Med. 6, 247ra103 (2014).
115. Mahu, I. & Domingos, A. I. The sympathetic neuro-​ of ATP and noradrenaline and their modulation by 167. Stefanidis, A. et al. Insights into the neurochemical
adipose connection and the control of body weight. neuropeptide Y in human vascular neuroeffector signature of the innervation of beige fat. Mol. Metab.
Exp. Cell Res. 360, 27–30 (2017). junctions. Eur. J. Pharmacol. 500, 27–35 (2004). 11, 47–58 (2018).
116. Dodd, G. T. & Tiganis, T. Insulin action in the brain: 142. Ralevic, V. & Dunn, W. R. Purinergic transmission in 168. Bernhard, F. et al. Functional relevance of genes
Roles in energy and glucose homeostasis. blood vessels. Auton. Neurosci. 191, 48–66 (2015). implicated by obesity genome-​wide association study
J. Neuroendocrinol. 29, e12513 (2017). 143. Razzoli, M. et al. Stress-​induced activation of brown signals for human adipocyte biology. Diabetologia 56,
117. Roh, E., Song, D. K. & Kim, M. S. Emerging role of adipose tissue prevents obesity in conditions of low 311–322 (2013).
the brain in the homeostatic regulation of energy and adaptive thermogenesis. Mol. Metab. 5, 19–33 169. Guilherme, A. et al. Adipocyte lipid synthesis coupled
glucose metabolism. Exp. Mol. Med. 48, e216 (2016). (2016). to neuronal control of thermogenic programming.
118. Benoit, S. C. et al. The catabolic action of insulin in the 144. Xie, T. R., Liu, C. F. & Kang, J. S. Sympathetic Mol. Metab. 6, 781–796 (2017).
brain is mediated by melanocortins. J. Neurosci. 22, transmitters control thermogenic efficacy of brown 170. Migrenne, S. et al. Fatty acid signaling in the
9048–9052 (2002). adipocytes by modulating mitochondrial complex V. hypothalamus and the neural control of insulin
119. Choudhury, A. I. et al. The role of insulin receptor Signal Transduct. Target. Ther. 2, 17060 (2017). secretion. Diabetes 55, 5 (2006).
substrate 2 in hypothalamic and beta cell function. 145. Pirzgalska, R. M. et al. Sympathetic neuron-​associated 171. Bazinet, R. P. & Laye, S. Polyunsaturated fatty acids
J. Clin. Invest. 115, 940–950 (2005). macrophages contribute to obesity by importing and their metabolites in brain function and disease.
120. Dodd, G. T. et al. Leptin and insulin act on POMC and metabolizing norepinephrine. Nat. Med. 23, Nat. Rev. Neurosci. 15, 771–785 (2014).
neurons to promote the browning of white fat. Cell 1309–1318 (2017). 172. Matias, I. et al. Regulation, function, and dysregulation
160, 88–104 (2015). 146. Tang, L. et al. Sympathetic nerve activity maintains of endocannabinoids in models of adipose and beta-​
121. Dodd, G. T. et al. A hypothalamic phosphatase an anti-​inflammatory state in adipose tissue in male pancreatic cells and in obesity and hyperglycemia.
switch coordinates energy expenditure with feeding. mice by inhibiting TNF-​alpha gene expression in J. Clin. Endocrinol. Metab. 91, 3171–3180 (2006).
Cell Metab. 26, 375–393 (2017). macrophages. Endocrinology 156, 3680–3694 173. Gonthier, M. P. et al. Identification of endocannabinoids
122. Shin, A. C. et al. Insulin receptor signaling in POMC, (2015). and related compounds in human fat cells. Obesity 15,
but not AgRP, neurons controls adipose tissue insulin 147. Camell, C. D. et al. Inflammasome-​driven catecholamine 837–845 (2007).
action. Diabetes 66, 1560–1571 (2017). catabolism in macrophages blunts lipolysis during 174. Matias, I. et al. Role and regulation of
123. Harris, R. B., Kelso, E. W., Flatt, W. P., Bartness, T. J. ageing. Nature 550, 119–123 (2017). acylethanolamides in energy balance: focus on
& Grill, H. J. Energy expenditure and body composition 148. Collins, S. Beta-​adrenoceptor signaling networks in adipocytes and beta-​cells. Br. J. Pharmacol. 152,
of chronically maintained decerebrate rats in the fed adipocytes for recruiting stored fat and energy 676–690 (2007).
and fasted condition. Endocrinology 147, 1365–1376 expenditure. Front. Endocrinol. 2, 102 (2011). 175. Ruiz de Azua, I. et al. Adipocyte cannabinoid receptor
(2006). 149. Zechner, R., Madeo, F. & Kratky, D. Cytosolic lipolysis CB1 regulates energy homeostasis and alternatively
124. Thaler, J. P. et al. Obesity is associated with and lipophagy: two sides of the same coin. Nat. Rev. activated macrophages. J. Clin. Invest. 127,
hypothalamic injury in rodents and humans. J. Clin. Mol. Cell Biol. 18, 671–684 (2017). 4148–4162 (2017).
Invest. 122, 153–162 (2012). 150. Granneman, J. G., Li, P., Zhu, Z. & Lu, Y. Metabolic and 176. Cote, M. et al. Circulating endocannabinoid levels,
125. Kleinridders, A. et al. MyD88 signaling in the CNS is cellular plasticity in white adipose tissue I: effects of abdominal adiposity and related cardiometabolic
required for development of fatty acid-​induced leptin beta3-adrenergic receptor activation. Am. J. Physiol. risk factors in obese men. Int. J. Obes. 31, 692–699
resistance and diet-​induced obesity. Cell Metab. 10, Endocrinol. Metab. 289, E608–E616 (2005). (2007).
249–259 (2009). 151. Ramseyer, V. D. & Granneman, J. G. Adrenergic 177. Di Marzo, V. et al. Changes in plasma endocannabinoid
126. Thaler, J. P. & Schwartz, M. W. Minireview: regulation of cellular plasticity in brown, beige/brite levels in viscerally obese men following a 1 year
inflammation and obesity pathogenesis: the and white adipose tissues. Adipocyte 5, 119–129 lifestyle modification programme and waist
hypothalamus heats up. Endocrinology 151, (2016). circumference reduction: associations with changes
4109–4115 (2010). 152. Bachman, E. S. et al. betaAR signaling required for in metabolic risk factors. Diabetologia 52, 213–217
127. Gao, Y. et al. Hormones and diet, but not body weight, diet-​induced thermogenesis and obesity resistance. (2009).
control hypothalamic microglial activity. Glia 62, Science 297, 843–845 (2002). 178. Wagner, I. V., Perwitz, N., Drenckhan, M., Lehnert, H.
17–25 (2014). 153. Douris, N. et al. Beta-​adrenergic receptors are critical & Klein, J. Cannabinoid type 1 receptor mediates
128. Garcia-​Caceres, C., Yi, C. X. & Tschop, M. H. for weight loss but not for other metabolic adaptations depot-​specific effects on differentiation, inflammation
Hypothalamic astrocytes in obesity. Endocrinol. to the consumption of a ketogenic diet in male mice. and oxidative metabolism in inguinal and epididymal
Metab. Clin. North Am. 42, 57–66 (2013). Mol. Metab. 6, 854–862 (2017). white adipocytes. Nutr. Diabetes 1, e16 (2011).

www.nature.com/nrendo
Reviews

179. Quarta, C. et al. CB(1) signaling in forebrain and 201. Collins, S., Daniel, K. W. & Rohlfs, E. M. Depressed 224. Zhao, Y. et al. Transient overexpression of VEGF-​A
sympathetic neurons is a key determinant of expression of adipocyte beta-​adrenergic receptors in adipose tissue promotes energy expenditure via
endocannabinoid actions on energy balance. is a common feature of congenital and diet-​induced activation of the sympathetic nervous system.
Cell Metab. 11, 273–285 (2010). obesity in rodents. Int. J. Obes. Relat. Metab. Disord. Mol. Cell. Biol. 38, e00242–18 (2018).
180. Bajzer, M. et al. Cannabinoid receptor 1 (CB1) 23, 669–677 (1999). 225. Jiang, Y., Berry, D. C. & Graff, J. M. Distinct cellular
antagonism enhances glucose utilisation and activates 202. Surwit, R. S., Dixon, T. M., Petro, A. E., Daniel, K. W. and molecular mechanisms for beta3 adrenergic
brown adipose tissue in diet-​induced obese mice. & Collins, S. Diazoxide restores beta3-adrenergic receptor-​induced beige adipocyte formation. eLife 6,
Diabetologia 54, 3121–3131 (2011). receptor function in diet-​induced obesity and diabetes. e30329 (2017).
181. Pakdeechote, P., Dunn, W. R. & Ralevic, V. Endocrinology 141, 3630–3637 (2000). 226. Ueta, C. B. et al. beta(1) Adrenergic receptor is
Cannabinoids inhibit noradrenergic and purinergic 203. Rahmouni, K. et al. Hypothalamic PI3K and MAPK key to cold- and diet-​induced thermogenesis in mice.
sympathetic cotransmission in the rat isolated differentially mediate regional sympathetic activation J. Endocrinol. 214, 359–365 (2012).
mesenteric arterial bed. Br. J. Pharmacol. 152, to insulin. J. Clin. Invest. 114, 652–658 (2004). 227. Shah, S. H. et al. Neuropeptide Y gene polymorphisms
725–733 (2007). 204. Muntzel, M. S., Morgan, D. A., Mark, A. L. confer risk of early-​onset atherosclerosis. PLOS Genet.
182. O’Keefe, L., Simcocks, A. C., Hryciw, D. H., Mathai, M. L. & Johnson, A. K. Intracerebroventricular insulin 5, e1000318 (2009).
& McAinch, A. J. The cannabinoid receptor 1 and produces nonuniform regional increases in 228. Sung, C. P., Arleth, A. J. & Feuerstein, G. Z.
its role in influencing peripheral metabolism. Diabetes sympathetic nerve activity. Am. J. Physiol. 267, Neuropeptide Y upregulates the adhesiveness of
Obes. Metab. 16, 294–304 (2014). R1350–R1355 (1994). human endothelial cells for leukocytes. Circ. Res. 68,
183. Pongratz, G. & Straub, R. H. The sympathetic nervous 205. Komohara, Y., Fujiwara, Y., Ohnishi, K., Shiraishi, D. 314–318 (1991).
response in inflammation. Arthritis Res. Ther. 16, 504 & Takeya, M. Contribution of macrophage polarization 229. Claxson, A. et al. The anti-​inflammatory effects of
(2014). to metabolic diseases. J. Atheroscler. Thromb. 23, D-​myo-inositol-1.2.6-trisphosphate (PP56) on animal
184. Xiong, Y. et al. Long-​acting MIC-1/GDF15 molecules 10–17 (2016). models of inflammation. Agents Act. 29, 68–70
to treat obesity: evidence from mice to monkeys. 206. Olefsky, J. M. & Glass, C. K. Macrophages, (1990).
Sci. Transl Med. 9, eaan8732 (2017). inflammation, and insulin resistance. Annu. Rev. 230. Singer, K. et al. Neuropeptide Y is produced by
185. Rines, A. K., Verdeguer, F. & Puigserver, P. Adenosine Physiol. 72, 219–246 (2010). adipose tissue macrophages and regulates obesity-​
activates thermogenic adipocytes. Cell Res. 25, 207. Lumeng, C. N., Bodzin, J. L. & Saltiel, A. R. Obesity induced inflammation. PLOS ONE 8, e57929 (2013).
155–156 (2015). induces a phenotypic switch in adipose tissue 231. Pandolfi, J. et al. Purinergic signaling modulates
186. Bootcov, M. R. et al. MIC-1, a novel macrophage macrophage polarization. J. Clin. Invest. 117, human visceral adipose inflammatory responses:
inhibitory cytokine, is a divergent member of the 175–184 (2007). implications in metabolically unhealthy obesity.
TGF-​beta superfamily. Proc. Natl Acad. Sci. USA 94, 208. Aron-​Wisnewsky, J. et al. Human adipose tissue J. Leukoc. Biol. 97, 941–949 (2015).
11514–11519 (1997). macrophages: m1 and m2 cell surface markers in 232. Eltzschig, H. K., Sitkovsky, M. V. & Robson, S. C.
187. Tsai, V. W., Lin, S., Brown, D. A., Salis, A. & Breit, S. N. subcutaneous and omental depots and after weight Purinergic signaling during inflammation. N. Engl.
Anorexia-​cachexia and obesity treatment may be two loss. J. Clin. Endocrinol. Metab. 94, 4619–4623 J. Med. 367, 2322–2333 (2012).
sides of the same coin: role of the TGF-​b superfamily (2009). 233. Jo, E. K., Kim, J. K., Shin, D. M. & Sasakawa, C.
cytokine MIC-1/GDF15. Int. J. Obes. 40, 193–197 209. Patsouris, D. et al. Ablation of CD11c-​positive cells Molecular mechanisms regulating NLRP3
(2016). normalizes insulin sensitivity in obese insulin resistant inflammasome activation. Cell. Mol. Immunol. 13,
188. Tsai, V. W. et al. Treatment with the TGF-​b superfamily animals. Cell Metab. 8, 301–309 (2008). 148–159 (2016).
cytokine MIC-1/GDF15 reduces the adiposity and 210. Oh, D. Y. et al. GPR120 is an omega-3 fatty acid 234. Enjyoji, K. et al. Deletion of cd39/entpd1 results in
corrects the metabolic dysfunction of mice with receptor mediating potent anti-​inflammatory and hepatic insulin resistance. Diabetes 57, 2311–2320
diet-​induced obesity. Int. J. Obes. 42, 561–571 insulin-​sensitizing effects. Cell 142, 687–698 (2010). (2008).
(2018). 211. Grailer, J. J., Haggadone, M. D., Sarma, J. V., 235. Chen, Z. et al. Nrg4 promotes fuel oxidation and a
189. O’Rahilly, S. GDF15 — from biomarker to allostatic Zetoune, F. S. & Ward, P. A. Induction of M2 regulatory healthy adipokine profile to ameliorate diet-​induced
hormone. Cell Metab. 26, 807–808 (2017). macrophages through the beta2-adrenergic receptor metabolic disorders. Mol. Metab. 6, 863–872 (2017).
190. Strelau, J., Schober, A., Sullivan, A., Schilling, L. with protection during endotoxemia and acute lung 236. Jeong, J. H., Chang, J. S. & Jo, Y. H. Intracellular
& Unsicker, K. Growth/differentiation factor-15 injury. J. Innate Immun. 6, 607–618 (2014). glycolysis in brown adipose tissue is essential for
(GDF-15), a novel member of the TGF-​beta superfamily, 212. Czech, M. P. Macrophages dispose of catecholamines optogenetically induced nonshivering thermogenesis
promotes survival of lesioned mesencephalic in adipose tissue. Nat. Med. 23, 1255–1257 (2017). in mice. Sci. Rep. 8, 6672 (2018).
dopaminergic neurons in vitro and in vivo and is 213. Cao, Y. Angiogenesis and vascular functions in 237. Akhmedov, D. et al. Gs-​DREADD knock-​in mice for
induced in neurons following cortical lesioning. modulation of obesity, adipose metabolism, and tissue-​specific, temporal stimulation of cyclic AMP
J. Neural Transm. Suppl. 65, 197–203 (2003). insulin sensitivity. Cell Metab. 18, 478–489 (2013). signaling. Mol. Cell. Biol. 37, e00584–16 (2017).
191. Gnad, T. et al. Adenosine activates brown adipose 214. Garg, J. et al. Catecholamines facilitate VEGF-​ 238. Rojas, J. M. & Schwartz, M. W. Control of hepatic
tissue and recruits beige adipocytes via A2A dependent angiogenesis via beta2-adrenoceptor-​ glucose metabolism by islet and brain. Diabetes Obes.
receptors. Nature 516, 395–399 (2014). induced Epac1 and PKA activation. Oncotarget 8, Metab. 16 (Suppl. 1), 33–40 (2014).
192. Dobson, J. G. Jr Mechanism of adenosine inhibition of 44732–44748 (2017). 239. Cantu, R. C. & Goodman, H. M. Effects of denervation
catecholamine-​induced responses in heart. Circ. Res. 215. Domigan, C. K. et al. Autocrine VEGF maintains and fasting on white adipose tissue. Am. J. Physiol.
52, 151–160 (1983). endothelial survival through regulation of metabolism 212, 207–212 (1967).
193. Rongen, G. A. et al. Presynaptic inhibition of and autophagy. J. Cell Sci. 128, 2236–2248 (2015). 240. Bartness, T. J., Shrestha, Y. B., Vaughan, C. H.,
norepinephrine release from sympathetic nerve 216. Lee, S. et al. Autocrine VEGF signaling is required for Schwartz, G. J. & Song, C. K. Sensory and sympathetic
endings by endogenous adenosine. Hypertension 27, vascular homeostasis. Cell 130, 691–703 (2007). nervous system control of white adipose tissue
933–938 (1996). 217. Marko, S. B. & Damon, D. H. VEGF promotes vascular lipolysis. Mol. Cell. Endocrinol. 318, 34–43 (2010).
194. Thorp, A. A. & Schlaich, M. P. Relevance of sympathetic innervation. Am. J. Physiol. Heart Circ.
sympathetic nervous system activation in obesity and Physiol. 294, H2646–H2652 (2008). Acknowledgements
metabolic syndrome. J. Diabetes Res. 2015, 341583 218. Long, J. B., Jay, S. M., Segal, S. S. & Madri, J. A. VEGF-​A Work from the laboratory of M.P.C. discussed in this Review
(2015). and semaphorin3A: modulators of vascular sympathetic was supported by NIH grants DK30898 and DK103047
195. Bougneres, P. et al. In vivo resistance of lipolysis to innervation. Dev. Biol. 334, 119–132 (2009). to M.P.C.
epinephrine. A new feature of childhood onset obesity. 219. Robciuc, M. R. et al. VEGFB/VEGFR1-induced
J. Clin. Invest. 99, 2568–2573 (1997). expansion of adipose vasculature counteracts obesity Author contributions
196. Horowitz, J. F. & Klein, S. Whole body and abdominal and related metabolic complications. Cell Metab. 23, A.G., F.H., A.H.B. and M.P.C. contributed equally to the dis-
lipolytic sensitivity to epinephrine is suppressed in 712–724 (2016). cussion of content, researching data for the article, writing
upper body obese women. Am. J. Physiol. Endocrinol. 220. During, M. J. et al. Adipose VEGF links the white-​ the article and reviewing and editing the manuscript before
Metab. 278, E1144–E1152 (2000). to-brown fat switch with environmental, genetic, and submission.
197. Heinonen, S. et al. Impaired mitochondrial biogenesis pharmacological stimuli in male mice. Endocrinology
in adipose tissue in acquired obesity. Diabetes 64, 156, 2059–2073 (2015). Competing interests
3135–3145 (2015). 221. Kim, K. H. et al. Intermittent fasting promotes The authors declare no competing interests.
198. Guo, T. et al. Adipocyte ALK7 links nutrient overload adipose thermogenesis and metabolic homeostasis via
to catecholamine resistance in obesity. eLife 3, VEGF-​mediated alternative activation of macrophage. Publisher’s note
e03245 (2014). Cell Res. 27, 1309–1326 (2017). Springer Nature remains neutral with regard to jurisdictional
199. Buettner, C. Is hyperinsulinemia required to develop 222. Xue, Y. et al. Hypoxia-​independent angiogenesis in claims in published maps and institutional affiliations.
overeating-​induced obesity? Cell Metab. 16, adipose tissues during cold acclimation. Cell Metab.
691–692 (2012). 9, 99–109 (2009). Reviewer information
200. Mehran, A. E. et al. Hyperinsulinemia drives diet-​ 223. Sun, K. et al. Brown adipose tissue derived VEGF-​A Nature Reviews Endocrinology thanks K. Rahmouni, and
induced obesity independently of brain insulin modulates cold tolerance and energy expenditure. other anonymous reviewers, for their contribution to the peer
production. Cell Metab. 16, 723–737 (2012). Mol. Metab. 3, 474–483 (2014). review of this work.

Nature Reviews | Endocrinology

Anda mungkin juga menyukai