Sei sulla pagina 1di 12

REVIEWS

The vagus nerve and the inflammatory


reflexlinking immunity and metabolism
Valentin A. Pavlov and Kevin J. Tracey
Abstract | The vagus nerve has an important role in regulation of metabolic homeostasis, and efferent
vagus nerve-mediated cholinergic signalling controls immune function and proinflammatory responses via
the inflammatory reflex. Dysregulation of metabolism and immune function in obesity are associated with
chronic inflammation, a critical step in the pathogenesis of insulin resistance and type2 diabetes mellitus.
Cholinergic mechanisms within the inflammatory reflex have, in the past 2years, been implicated in attenuating
obesity-related inflammation and metabolic complications. This knowledge has led to the exploration of novel
therapeutic approaches in the treatment of obesity-related disorders.
Pavlov, V. A. & Tracey, K. J. Nat. Rev. Endocrinol. 8, 743754 (2012); doi:10.1038/nrendo.2012.189

Introduction
The vagus nerve (cranial nerve X) is the main nerve of Proinflammatory progression is balanced by the release
the parasympathetic division of the autonomic nervous of IL-10, TGF, soluble cytokine receptors and other
system. The vagus nerve regulates metabolic homeostasis anti-inflammatory molecules.
by controlling heart rate, gastrointestinal motility and Inflammation is normally a local and temporary
secretion, pancreatic endocrine and exocrine secretion, event and, upon its resolution, immune and physio
hepatic glucose production, and other visceral functions. logical homeostasis is restored. However, disrupted
In addition, the vagus nerve is a major constituent of a innate immune regulation can result in continual pro
neural reflex mechanismthe inflammatory reflexthat inflammatory cytokine activity and excessive or chronic
controls innate immune responses and inflammation inflammation. This state underlies the pathogenesis of a
during pathogen invasion and tissue injury.13 range of disease syndromes, including sepsis, rheumatoid
Innate immune responses are activated by pathogen- arthritis, inflammatory bowel disease and other inflam-
associated and danger-associated molecular patterns matory and autoimmune disorders.810 Understanding
that are recognized by sensors on the immune cell endogenous mechanisms that prevent or neutralize exces-
surface or in intracellular compartments. These cellular sive proinflammatory responses could lead to novel thera
sensors include Toll-like receptors (TLRs), nucleotide- peutic options for diseases associated with an excessive or
binding oligomerization domain-like receptors (NLRs) chronic inflammatory state.
and other pattern-recognition receptors (Figure 1).46 Chronic inflammation as a result of immune and meta
Activation of signalling cascades downstream of TLRs bolic dysregulation is a characteristic feature in patients
results in increased production and release of tumour with obesity and is causally linked with insulin resist-
necrosis factor (TNF), IL6 and other proinflammatory ance and other metabolic complications.1113 Decreased
cytokines.1,7 In addition, activation of NLRs is associated vagus nerve activity in the context of obesity has been
with the formation of multimeric protein complexes, reported.1417 Selective cholinergic activation within the
termed inflammasomes, which regulate maturation efferent vagus nerve-mediated arm of the inflammatory
and release of the proinflammatory cytokines IL1 and reflex can suppress obesity-associated inflammation
IL18.5 Proinflammatory cytokines, along with chemo and reverse metabolic complications. 1820 These find-
kines, reactive oxygen species, nitrogen intermediates ings raise the intriguing possibility that dysregulation of
and other inflammatory molecules, are critically impli- vagus nerve-mediated signalling might contribute to the
cated in extracellular pathogen clearance, vasodilatation, pathogenesis of obesity and its related comorbidities.
neutrophil recruitment, increased vascular perme- In this Review, we provide a conceptual view of the Center for Biomedical
ability and induction of acute-phase proteins, such as inflammatory reflex as a physiological mechanism that Science, The Feinstein
Institute for Medical
Creactive protein (CRP), and coagulation molecules.1,5,7 functions on the path between immunity and metabo- Research, 350
lism and could be exploited in the treatment of obesity- Community Drive,
associated inflammation and obesity-related disorders. Manhasset, NY 11030,
USA (V. A. Pavlov,
Competing interests K.J.Tracey).
The authors declare that they are inventors on patents related to The inflammatory reflex
the content of this manuscript.K. J. Tracey also declares an Correspondence to:
association with the following company: SetPoint Medical. See Communication between the immune system and V. A. Pavlov
the article online for the full details of the relationships. the brain is vital for controlling inflammation. The vpavlov@nshs.edu

NATURE REVIEWS | ENDOCRINOLOGY VOLUME 8 | DECEMBER 2012 | 743


2012 Macmillan Publishers Limited. All rights reserved
REVIEWS

Key points organs (including the area postrema) and via other
humoral mechanisms (Figure 1). 28,29 Accordingly,
The inflammatory reflex is a physiological mechanism through which the vagus
nerve regulates immune function and inhibits excessive proinflammatory afferent vagus nerve endings appear to be important
cytokine production for relaying information about immune status to the
Vagus nerve signalling has an important role in the regulation of feeding brain when proinflammatory cytokines are present at
behaviour and metabolic homeostasis fairly low levels.24,28 However, TLR4 (which can be acti-
Disruption of metabolic and immune regulation in obesity results in vated by lipopolysaccharide) is expressed in the nodose
inflammation, which mediates insulin resistance and the development of type2 ganglion, 30,31 which suggests a mechanism by which
diabetes mellitus as well as other debilitating and life-threatening conditions
lipopolysaccharide and other inflammatory molecules
Activation of cholinergic signalling in the efferent arm of the inflammatory reflex
alleviates obesity-associated inflammation and metabolic derangements
can activate vagus nerve afferents above their visceral
The inflammatory reflex can potentially be exploited for treatment of the endings (Figure1).
metabolic syndrome, type2 diabetes mellitus and other obesity-driven disorders
Efferent arm
A little more than a decade ago, an important role of
inflammatory reflex is a centrally integrated physiological efferent vagus nerve cholinergic signalling in brain-to-
mechanism in which afferent vagus nerve signalling, acti- immune communication was revealed by observations
vated by cytokines or pathogen-derived products, is func- that vagus nerve stimulation suppresses local and serum
tionally associated with efferent vagus nerve-mediated proinflammatory cytokine levels in rodents with endo
output to regulate proinflammatory cytokine production toxaemia, and that acetylcholine inhibits the release
and inflammation (Figure1). The absence of this inflam- of TNF, IL1 and IL18 from lipopolysaccharide-
matory reflexresulting from neural lesions or genetic stimulated macrophages. 21 These findings led to the
ablation of essential componentsresults in excessive definition of the cholinergic anti-inflammatory pathway
innate immune responses and cytokine toxicity.21,22 as the efferent vagus nerve-based arm of the inflam-
The inflammatory reflex has been comprehensively matory reflex (Figure1). 21 This efferent arm can be
reviewedelsewhere.13,9,23 centrally regulated, and muscarinic acetylcholine recep-
tors in the brain have been implicated in this regula-
Afferent arm tion (Figure1).3235 Galantamine is a centrally acting
Afferent vagus nerve fibres sense peripheral inflamma- acetylcholinesterase inhibitor and activates the efferent
tory molecules and convey signals to the brain and are, cholinergic arm of the inflammatory reflex by through
therefore, important for immune-to-brain communica- a muscarinic receptor-dependent mechanism in the
tion (Figure1).1,24 Afferent vagus neurons, residing in the brain.34 Defining a brain network that can be used to
nodose and jugular ganglia, terminate primarily in explore central mechanisms of inflammatory reflex
the nucleus tractus solitarius in the brainstem medulla control is the aim of ongoing studies.
oblongata (Figure1).24 Afferent signalling is further In peripheral tissues, the 7 nicotinic acetylcholine
communicated through neural contacts between brain- receptor (7nAChR) is important for mediating anti-
stem nuclei, the hypothalamus and forebrain regions inflammatory signalling within the efferent arm of the
associated with integration of visceral sensory informa- inflammatory reflex (Figure2).22 Accordingly, several
tion as well as coordination of autonomic function and 7nAChR agonists have been identified as experimental
behavioural responses.23,24 anti-inflammatory therapeutics with potential for clini-
Peripheral administration of bacterial lipopolysaccha- cal development.2,3638 The 7 subunit of the receptor is
ride (also known as endotoxin) or IL1 causes afferent expressed in macrophages, monocytes, dendritic cells,
vagus nerve activation, as determined by increased cFos Tcells, endothelial and other non-neuronal cells, and
expression and electrical activity. 24,25 IL1 receptors the structural and functional characterization of non-
expressed on vagus nerve afferents and chemosensory neuronal 7nAChRs is an area of ongoing study.2,38 The
(glomus) cells in paraganglia surrounding afferent vagus presence of 7nAChRs on bone marrow-derived cells is
nerve endings have been implicated in the recognition required for the functional integrity of the inflamma-
of immune activation (Figure1).24,26,27 Prostaglandin- tory reflex, but their presence on Tcells and neuronal
dependent mechanisms have also been implicated in cells is not essential.39 The anti-inflammatory action of
activating vagus nerve afferents by increasing levels 7nAChR agonists or vagus nerve stimulation is associ-
of circulating IL1. 26 Intraportal administration of ated with downregulation of CD14 and TLR4 expression
IL1 to rats increases afferent and, subsequently, effer- in immune cells.40 Cholinergic inhibition of proinflam-
ent vagus nerve and splenic nerve activity.25 Peripheral matory cytokine production is mediated through intra-
immune stimulation causes sickness behaviour, which cellular signal pathways downstream of 7nAChR that
is attenuated in rodents who have undergone surgical culminate in suppression of NFB nuclear transloca-
transection of the vagus nerve to prevent immune sig- tion (Figure2).3638,4042 The tyrosine protein kinase JAK2
nalling at peripheral nerve endings.24 This attenuation might also be recruited and activated by 7nAChR upon
is dependent on the magnitude of immune activation, cholinergic stimulation.43 Subsequent phosphorylation of
because fairly large quantities of circulating IL1 the transcription factor STAT3 results in suppression
produce fever and sickness behaviour by bypassing the of proinflammatory cytokine production in intestinal
neural circuits, acting through brain circumventricular macrophages (Figure 2).43

744 | DECEMBER 2012 | VOLUME 8  www.nature.com/nrendo


2012 Macmillan Publishers Limited. All rights reserved
REVIEWS

The anti-inflammatory effects of vagus nerve stimula-


tion in animals with endotoxaemia require neural signals mAChR AChE
along the adrenergic splenic nerve,44 which originates ligands inhibitors

in the celiac ganglion that is innervated by the efferent


vagus nerve (Figure1, Figure2). Somewhat unexpect- mAChR

edly, a specific subset of Tcells (memory CD4+ Tcells,


which express adrenoreceptors), instead of neurons,
provide an endogenous source of acetylcholine in the
spleen (Figure2).45 Tcell production of acetylcholine
in the spleen is critical for the inflammatory reflex. In NTS
DMN
nude mice, which lack Tcells, vagus nerve stimula-
tion fails to suppress TNF levels during endotoxaemia. NA AP
However, transfer of acetylcholine-producing Tcells,
which repopulate the spleen in nude mice, restores the
integrity of the neural circuit.45 In addition to this spleen- Efferent Nodose
mediated mechanism in endotoxaemia, efferent vagus vagus ganglion
nerve
nerve endings might directly regulate the immune func- Heart
tion by releasing acetylcholine, without the requirement TLR4
for either signalling along the splenic nerve or Tcells
(Figure1, Figure2). Such a direct mechanism has been
implicated in the suppression of inflammation in experi-
mental settings of haemorrhagic shock, ileus, inflamma-
Liver
tory bowel disease, autoimmune myocarditis and other
Celiac
inflammatory and autoimmune conditions.2,10,41,43,4649 ganglion
Importantly, many of these disorders are associated with
autonomic dysfunction and decreased vagus nerve tone.
Enhancement of vagus nerve output could, therefore, Afferent
have therapeutic potential in these settings.50,51 Splenic
vagus
nerve
Gastrointestinal nerve
tract
Vagus nerve in metabolic regulation
Vagus nerve afferent and efferent signalling has an
LPS, other
important role in the regulation of feeding behaviour Spleen pathogen fragments
and metabolic homeostasis. This finely tuned regula- and tissue-
injury molecules
tion is aimed at preserving energy balance and prevent- Chemo-
sensory
ing fluctuations in body weight and metabolism that can paraganglia
Intestine
be detrimental to the individual.

Dietary intake and metabolism IL-1R TLRs


Vagus nerve afferents innervating the gastrointestinal
NLRs
tract and liver are major constituents of a sensory system
that detects changes in micronutrient and metabolic mol- Proinflammatory
cytokines
ecules. These nerve fibres transmit information detected
by associated mechanoreceptors, chemoreceptors and
Macrophages,
specific metabolite receptors in the gut and hepatic dendritic cells,
portal system concerning levels of lipids, cholecysto- other immune cells
kinin, leptin, peptide YY, insulin and glucose to the brain
(Figure3).52,53 Vagus nerve efferents, on the other hand, Figure 1 | The functional anatomy of the inflammatory reflex. Inflammatory
provide brain-derived output to the gastrointestinal tract, mediators, such as cytokines, are released by activated macrophages and other
immune cells when TLRs and NLRs are activated upon immune challenge. These
liver and pancreas (Figure3).52 Vagus nerve innerva-
mediators are detected by sensory components of the afferent arm of the
tion of white adipose tissue has also been demonstrated inflammatory reflex (red). Neuronal interconnections between the NTS, AP, DMN,
using retrograde neuronal tracing.54 Other researchers, NA, and higher forebrain regions (not shown) integrate afferent signalling and
however, found limited retrograde labelling in the dorsal efferent vagus nerve-mediated immunoregulatory output. Efferent vagus nerve
motor nucleus following injection of the tracer into cholinergic output to the spleen, liver and gastrointestinal tract (blue) regulates
white adipose tissue and a lack of some parasympathetic immune activation and suppresses proinflammatory cytokine release (dotted red
markers in adipose tissue.55 Therefore, the anatomy and lines). This efferent cholinergic arm of the inflammatory reflex can be activated in
a functional role of the vagus nerve in relation to adipose the brain through mAChR-mediated mechanisms triggered by mAChR ligands and
AChE inhibitors, such as galantamine. Abbreviations: AChE, acetylcholinesterase;
tissue is an area of active study.
AP, area postrema; DMN, dorsal motor nucleus of the vagus nerve; LPS,
Hepatic and gastrointestinal vagus nerve afferents lipopolysaccharide (endotoxin); mAChR, muscarinic acetylcholine receptor; NA,
are involved in the regulation of short-term feeding nucleus ambiguus; NLRs, nucleotide-binding oligomerization domain-like
behaviour.52,53 Afferent vagus nerve signalling mediates receptors; NTS, nucleus tractus solitarius; TLR4, toll-like receptor 4.

NATURE REVIEWS | ENDOCRINOLOGY VOLUME 8 | DECEMBER 2012 | 745


2012 Macmillan Publishers Limited. All rights reserved
REVIEWS

Celiac ganglion the lumen of the gastrointestinal tract causes neuronal


activation in vagus nerve afferents, the nucleus tractus
Splenic Efferent vagus
nerve nerve solitarius, the arcuate hypothalamic nucleus and the
dorsal motor nucleus, thus highlighting the role of brain
Spleen circuitry in vagus nerve regulation of gastrointestinal
NA
function and pancreatic secretion.61
Additionally, metabolic molecules can act directly in
the brain to trigger efferent output that regulates meta-
2AR
bolic homeostasis (Figure3). Insulin signalling in the
mediobasal hypothalamus is mediated through modula-
T cell tion of ATP-sensitive potassium channels and has been
implicated in the suppression of hepatic glucose pro-
duction.62,63 Efferent vagus nerve signalling to the liver
Liver
Intestine is essential for this regulation.63 Hypothalamic sensing
of fluctuations in circulatory lipid levels triggers effer-
ent vagus nerve-mediated output that regulates hepatic
Efferent vagus
nerve Efferent vagus glucose production and glucose homeostasis.64 In the
ACh nerve hypothalamus, cholinergic activation dependent on
muscarinic receptors increases hepatic glycogen synthe-
sis through a mechanism mediated by the vagus nerve.65,66
Vagus nerve-derived cholinergic signalling through a
mechanism mediated by M3 muscarinic receptors has
7nAChR
7nAChR also been implicated in stimulating insulin release in
JAK2 thepancreas.67
Macrophages, P P STAT3
The dorsal vagal complex and different hypothalamic
dendritic cells,
other immune cells P P regions are important constituents of a brain network
associated with vagus nerve-mediated regulation
NF-B
of peripheral metabolic functions (Figure3). Inter
connections between these and other forebrain regions
also have a role in the integration of afferent and efferent
signalling in terms of the control of metabolic homeo-
stasis and the regulation of hedonic and motivational
Proinflammatory cytokines aspects of feeding behaviour (Figure3).6870
Figure 2 | Molecular mechanisms of cholinergic control of inflammation. Efferent
vagus nerve activity is translated into catecholamine-mediated activation of Tcell- Postprandial inflammation
derived acetylcholine release in the spleen and into direct acetylcholine release Serum lipopolysaccharide levels in healthy individuals
from efferent vagus nerve endings in other organs. Inhibition of NFB nuclear are increased after consuming high-fat, or high-fat and
translocation and activation of a JAK2STAT3-mediated signalling cascade in high-carbohydrate meals.71,72 Endotoxaemia following
macrophages and other immune cells are implicated in cholinergic 7nAChR- high-fat diet ingestion or lipid intake has been implicated
mediated control of proinflammatory cytokine production. Abbreviations: ACh, in postprandial inflammation as plasma levels of IL6
acetylcholine; 2AR, 2 adrenergic receptor; JAK2, Janus kinase 2; 7nAChR, 7
and the expression of TLR4, TLR2 and SOCS3 in mono-
nicotinic acetylcholine receptor; NA, noradrenaline; NFB, nuclear factor B;
STAT3, signal transducer and activator of transcription 3. nuclear cells increases.7274 Postprandial endotoxaemia
and inflammation are transient, however, and data from
several studies suggest that the vagus nerve and the
gastric cholecystokinin-induced satiety and meal ter- inflammatory reflex might have a role in suppressing
mination.52 Synergistic activation of vagus nerve affer- postprandial inflammation. Afferent vagus nerve sig-
ents by both cholecystokinin and leptin mediates their nalling has a primary role in informing the brain about
short-term inhibitory effects on food intake. 53 Lipid the presence of peripheral inflammation during expo-
accumulation in the upper intestine, and consequent sure to low levels of proinflammatory stimuli.28 TLR4,
intestinal cholec ystokinin release, also triggers vagus which is expressed by vagus nerve afferents,30,31 provides
nerve-mediated and brain-integrated suppression of a molecular sensory component for neural detection of
hepatic glucose production.56,57 Additionally, the efferent lipopolysaccharide in postprandial inflammation.
arm of this reflex mechanism can be stimulated by direct Dietary lipid infusion causes the release of chole
activation of neurons in the dorsal vagal complex.58 cystokinin, which acts both via vagus nerve afferents and
Glucose-induced pancreatic insulin secretion can directly in the brain to trigger activation of efferent vagus
be stimulated by treatment with a truncated form of nerve signalling, which in turn suppresses the release of
glucagon-like peptide1, which activates hepatic vagus proinflammatory cytokines.75 Activation of vagus nerve
nerve afferents and, subsequently, vagus nerve efferents afferents by cholecystokinin or leptin is potentiated by
innervating the pancreas.59,60 In addition to its intrin- IL1.76,77 Furthermore, in rats, truncal vagotomy is asso-
sic effects on the enteric nervous system, glucose in ciated with increased bacterial translocation across the

746 | DECEMBER 2012 | VOLUME 8  www.nature.com/nrendo


2012 Macmillan Publishers Limited. All rights reserved
REVIEWS

intestinal mucosa,78 which suggests a tonic vagus nerve


control of intestinal permeability and postprandial
Cortex and other forebrain structures
endotoxaemia.

The inflammatory reflex and obesity Hedonic aspects of feeding behaviour


Disruption in metabolic and immune homeostasis in
obesity is associated with hyperglycaemia, insulin resist- Hypothalamus
ance, dyslipidaemia and hypertension. This cluster of
conditions characterizes the metabolic syndrome. 79
Moreover, levels of proinflammatory cytokines and Satiety

acute-phase proteins such as CRP are increased in indivi


duals with obesity, indicating chronic inflammation.11,80 AP
This inflammatory state is considered to be an essential
NTS DMN
pathophysiological constituent in obesity, underlying its
adverse consequences and linking it to the other compo-
nents of the metabolic syndrome.80,81 Several lines of evi-
dence indicate that vagus nerve activity could be impaired Afferent Efferent
in obesity, and enhancing cholinergic signalling within vagus vagus
nerve nerve
the inflammatory reflex can suppress obesity-associated
inflammation and its adverse implications.

Inflammation and obesity pathogenesis


The characteristic inflammatory state in obesity has CCK
Regulation of:
been extensively discussed elsewhere.11,13,80,82,83 White Leptin GLP-1 Gastrointestinal motor
adipose tissue in individuals with obesity is expanded and secretory functions
Lipids PYY Gastrointestinal tract Hepatic glucose production
and infiltrated with Tcells, bone-marrow-derived and hepatic portal Glycogen synthesis
macrophages, mast cells and Bcells.83 As such, this tissue Glucose system Pancreatic endocrine (insulin)
and exocrine secretion
becomes a major source of proinflammatory factors.80,8386
Adipocyte enlargement as a result of increased lipid Figure 3 | The role of the vagus nerve in metabolic regulation. Gastrointestinal and
deposition leads to metabolic alterations associated with hepatic vagus nerve afferents (red) communicate alterations in peripheral levels of
dysregulated secretion of adipokines, including leptin, micronutrients and metabolic molecules to the brain. Neural interaction between
resistin, adiponectin and visfatin.80,84 Additionally, cell the interconnected NTS, DMN and AP, within the dorsal vagal complex, and
death and hypoxia occur in adipose tissue, leading to reciprocal projections between this brainstem region and several hypothalamic
increased macrophage recruitment and generation of areas (arcuate and paraventricular nuclei and mediobasal and lateral areas),
underlie brain integration of visceral information and the modulation of efferent
reactive oxygen species, which promote inflammation.85,87
motor vagus nerve output, leading to regulation of metabolic homeostasis.
Increased production and release of TNF, IL6, CCL2 and Efferent vagus nerve signalling (blue) can be triggered by sensing metabolic
other proinflammatory mediators from adipocytes, as well alterations in the brainstem and the hypothalamus. Complex communication
as from activated M1 macrophages and other immune between hypothalamic nuclei and other forebrain structures (such as the insula
cells, also drives inflammation.80,82,84,85 In addition to and premotor cortex, amygdala, nucleus accumbens, parabrachial nucleus and
adipose tissue, activation of inflammatory pathways also thalamus) mediate hedonic, motivational and rewarding aspects of feeding
occurs in the liver, skeletal muscle and brain.12,82 behaviour and their interaction with vagus nerve-mediated homeostatic
Adipocyte and macrophage expression of TLR4 and mechanisms. Abbreviations: AP, area postrema; CCK, cholecystokinin; DMN,
dorsal motor nucleus of the vagus nerve; GLP1, glucagon-like peptide1;
TLR2 provides a mechanism for transforming metabolic
NA, nucleus ambiguus; NTS, nucleus tractus solitarius; PYY, peptide YY.
overload (high levels of saturated free fatty acids and
glucose) into proinflammatory responses, as free fatty
acids and glucose can be ligands for TLRs. Subsequent obesity.91,92 Chronic ingestion of a high-fat diet increases
intracellular signalling mediated through JNK, IKK and the proportion of lipopolysaccharide-containing micro-
PKR leads to stimulation of the transcription factors biota in the gut and this increase is associated with
NFB and AP1, and increased production of pro low-grade metabolic endotoxaemia in both mice and
inflammatory mediators.82,84,85,88 Metabolic overload also humans.92,93 In mice, mimicking metabolic endotoxae-
results in endoplasmic reticulum stress, which is asso- mia by chronic administration of a low dose of lipopoly-
ciated with JNK and IKK signalling and generation of saccharide results in inflammation, increased adiposity,
reactive oxygen species, triggering inflammation.13,82 In weight gain and metabolic complications.93 These changes
addition, activation of macrophages and Tcells in the result from activation of CD14TLR4 signalling, which
adipose tissue of patients with obesity can be mediated triggers increased production of proinflammatory
by inflammasomes.89 Pancreatic islet inflammation, as cytokines by adipocytes and macrophages.85,93
occurs in patients with type2 diabetes mellitus, can also The role of specific cytokines and adipokines in medi-
be mediated by these protein complexes.90 ating obesity-associated complications (Table1) has been
The intestinal microflora also contribute to the the subject of several reviews.83,84,88,9496 Inflammation
developm ent of inflammation in individuals with is causally linked with impaired insulin signalling in

NATURE REVIEWS | ENDOCRINOLOGY VOLUME 8 | DECEMBER 2012 | 747


2012 Macmillan Publishers Limited. All rights reserved
REVIEWS

Table 1 | Contributions of key adipokines and cytokines to obesity-related disease


Cytokines and adipokines Cell source Role in obesity pathogenesis
Leptin (adipokine) Adipocytes Regulates food intake and energy balance136*
Gastric mucosa cells Increased release in parallel with weight gain*
Activates anorexigenic pathways in the hypothalamic arcuate nucleus and
other brain regions69,136*
Acts through leptin receptors on vagus nerve afferents and on brain neurons
and via intracellular JAK2STAT3-mediated signalling31,69,136*
Positive regulator of glucose homeostasis and insulin sensitivity in muscle
and liver136*
Leptin resistance in obesity associated with intracellular SOCS3-mediated
suppression of STAT3 phosphorylation98,136
Hyperleptinaemia has proinflammatory effects in obesity84,137,138
TNF (proinflammatory Macrophages Major contributor to local and systemic insulin resistance80,82,85,139
cytokine) Other immune cells Increased expression in adipose tissue139
Adipocytes Stimulates resistin expression in human macrophages and suppresses
adiponectin expression in human adipocytes83,88
Implicated in pathogenesis of fatty liver disease94
IL6 (proinflammatory Macrophages Mediates local and systemic insulin resistance80,82,85
cytokine) Other immune cells Suppresses adiponectin expression in human adipocytes
Adipocytes Implicated in pathogenesis of fatty liver disease94
Inducer of acute-phase proteins, including C-reactive protein in liver, which is
linked to an increased risk of cardiovascular disease80,96,140
Increased expression in adipose tissue and increased circulating levels
in obesity13,80
CCL2 (chemokine) Adipocytes Mediates macrophage and other immune cell infiltration into adipose tissue141
Endothelial cells Linked to insulin resistance and hepatosteatosis in rodents141
Macrophages Increased levels in obesity85
Resistin (adipokine) Adipocytes (mice) Induces insulin resistance in mice, and might have a similar role
Macrophages in humans85,142
(humans) Stimulates TNF and IL6 production84
Increased circulating and adipose tissue levels in rodent models of obesity11
Adiponectin (adipokine) Adipocytes Anti-inflammatory and insulin-sensitizing functions95
Suppresses macrophage activation and proinflammatory cytokine release84
Inversely correlated with insulin resistance, type2 diabetes mellitus, fatty
liver disease and atherosclerosis84,94
Decreased circulating levels in obesity84
*Physiological functions under normal conditions. Abbreviations: CCL2, CCmotif chemokine 2 (also known as monocyte chemotactic protein 1 or MCP1); TNF,
tumour necrosis factor.

peripheral target tissues and insulin resistance, a major Vagus nerve signalling in obesity
complication in obesity and type2 diabetes melli- Obesity and obesity-related type2 diabetes mellitus are
tus.12,84,85,95 Activation of inflammatory pathways in the associated with attenuation of the afferent and effer-
brain interferes with insulin and leptin signalling in ent vagus nerve signalling that is implicated in meta-
the brain and also contributes to insulin resistance.82,97 bolic regulation. Obesity in rodents fed a high-fat diet
Increased lipolysis in insulin-resistant adipose tissue leads results in leptin resistance (indicated by decreased
to enhanced release of free fatty acids, which stimulate leptin-mediated STAT3 phosphorylation) in afferent
inflammatory responses and reinforce insulin resist- vagus neurons.31 This decrease occurs in parallel with an
ance. Proinflammatory cytokines and adipokines can increase in expression of SOCS3 (an inhibitor of STAT3
inhibit insulin signalling by targeting IRS proteins or phosphorylation) in nodose ganglia.31 SOCS3 expression
the insulin receptor. Specifically, TNF induces inhibi- is also induced by lipopolysaccharide. Consequently,
tion of insulin-activated tyrosine phosphorylation of the endotoxaemia, resulting from impaired gut perme
insulin receptor and serine phosphorylation of IRS1, ability, 99 could also contribute to leptin resistance
which leads to inactivation of insulin signalling.85,95 TNF in vagus nerve afferents through activation of TLR4.31 In
and other proinflammatory cytokines act in a paracrine rodents, the progression of obesity-related type2 diabetes
manner to stimulate JNK, IKK and possibly other kinases, mellitus is associated with attenuation of glucose-induced
which mediate serine phosphorylation of IRS proteins.82 afferent vagus nerve signalling.100
Additionally, proinflammatory cytokines such as TNF, Experimental evidence indicates that vagus nerve sig-
IL6 and the adipokine resistin induce the expression of nalling regulating hepatic glucose production, a major
SOCS proteins, which block insulin signalling through determinant of blood glucose levels, is decreased in
inhibition of insulin receptor tyrosine kinase activity or obesity. For instance, a high-fat diet causes impairment of
IRS1 ubiquitination and degradation.95,98 The net result a cholecystokinin-triggered vagus nerve reflex circuit that
is a negative influence of proinflammatory cytokines on suppresses hepatic glucose production.57 Additionally, the
insulin signalling. fatty acid-triggered hypothalamic suppression of hepatic

748 | DECEMBER 2012 | VOLUME 8  www.nature.com/nrendo


2012 Macmillan Publishers Limited. All rights reserved
REVIEWS

glucose production, which is vagus nerve-mediated,101 is marker F4/80, indicating that macrophage infiltration and
attenuated by voluntary overeating.102 This attenuation inflammation were bothsuppressed.19
has been suggested to contribute to the rapid onset of Mice lacking 7nAChR that were fed a high-fat diet
weight gain and hepatic insulin resistance, 102 and has had greater M1 macrophage infiltration and higher
been postulated as a feature of obesity-related type2 expression of TNF and CCL2 in adipose tissue than
diabetes mellitus.101 was observed in wild-type mice.19 Additionally, expres-
Insulin resistance in sucrose-fed rats is associated sion of proinflammatory genes following exposure to
with impaired muscarinic receptor-mediated hepatic free fatty acids or TNF was higher in macrophages from
vagus nerve signalling,103 as well as impaired baroreflex mice lacking 7nAChR than in macrophages from wild-
sensitivity resulting from decreased vagus nerve activ- type mice.19 Nicotine treatment failed to suppress this
ity.104 Vagus nerve cholinergic and muscarinic receptor- increased expression of proinflammatory genes in macro
dependent signalling controls glucose-stimulated insulin phages from mice lacking the receptor.19 These results
secretion by pancreatic cells, and mice with selective provide a mechanistic link between the inflammatory
deficiency of the M3muscarinic acetylcholine receptor reflex and signalling through 7nAChR in the inhibition
in pancreatic cells have suppressed insulin secretion and of obesity-associated inflammation.
impaired glucose tolerance.105 Furthermore, a high-fat diet One pharmacological strategy to activate the efferent
fails to induce glucose intolerance and hyperglycaemia in cholinergic arm of the inflammatory reflex is adminis
transgenic mice with selective overexpression of the M3 tration of the centrally-acting acetylcholinesterase inhib-
muscarinic receptor on pancreatic cells.105 itor galantamine.34 This compound enhances cholinergic
Autonomic dysfunction and diminished vagus nerve muscarinic receptor-mediated signalling in the brain and
activity occur frequently in individuals with obesity and stimulates efferent vagus nerve activity.34,108 Galantamine
type2 diabetes mellitus.1416 A 15-year follow-up study has treatment of mice with high-fat-diet-induced obesity and
revealed a strong relationship between autonomic dys- hyperglycaemia suppressed plasma IL6, CCL2, leptin
function and insufficient vagus nerve activity (revealed and resistin levels.20 Importantly, galantamine treatment
by impaired heart rate recovery following exercise cessa- of obese mice lowered plasma IL6 and CCL2 expres-
tion), impaired glucose homeostasis and development of sion to levels detected in lean control mice.20 Collectively,
type2 diabetes mellitus.17 Together, these preclinical and these findings highlight that activation of components of
clinical findings support the hypothesis that diminished the inflammatory reflex results in inhibition of obesity-
vagus nerve signalling in obesity could lead to enhanced associated inflammation.
inflammation and metabolic complications.
Alleviation of metabolic complications
Cholinergic alleviation of obesity Nicotine administration to mice fed a high-fat diet signif-
Targeting cholinergic mechanisms in the inflamma- icantly improves glucose homeostasis and insulin signal-
tory reflex using 7nAChR agonists or a centrally- ling by restoring tyrosine phosphorylation of the insulin
acting acetylcholinesterase inhibitor could alleviate receptor and IRS1 in skeletal muscle, adipose tissue and
inflammation and metabolic complications in obesity. liver.19 Administration of the 7nAChR agonist TC7020
to Leprdb/db mice reduces weight gain, food intake and
Attenuation of inflammation levels of blood glucose, HbA1c and triglycerides.18 When
Murine adipocytes express 7nAChR, and treatment this treatment is combined with administration of the
with the receptor agonist nicotine suppresses adipocyte selective 7nAChR antagonist methyllycaconitine,
TNF levels.19,106 This receptor is also expressed in human these effects are abolished.18 In contrast to the effects
adipocytes and has a role in suppressing proinflammatory of TC7020 administration, treatment with a selective
gene expression.107 Oral administration of the selective agonist targeting 4/2nAChR, which is predominantly
7nAChR agonist TC7020 to genetically obese Leprdb/db expressed on brain neurons, suppresses only weight
mice (which lack functional leptin receptors) significantly gain and food intake. 18 Administration of an inhibi-
suppresses serum TNF levels. This effect is abolished by tor of JAK2 counters the effects of TC7020 treatment
co-administration of the selective 7nAChR antago- on body weight, food intake and blood glucose levels,
nist methyllycaconitine.18 Administration of a selective thus indicating a mechanistic link between 7nAChR
4/2nAChR agonist failed to suppress serum TNF levels and JAK2-mediated signalling. Mice lacking 7nAChR
in Leprdb/db mice, which indicates the 7nAChR-specific that are fed a high-fat diet have greater impairment in
nature of this effect.18 Administration of nicotine to Leprdb/db glucose homeostasis, insulin sensitivity, and insulin sig-
mice and mice with high-fat-diet-induced obesityin nalling in muscle and liver than do wild-type mice fed
doses that did not affect food intake and body weight the same diet.19 In addition, the expression of human
significantly ameliorated inflammation in adipose tissue adipocyte 7nAChR (which suppresses proinflammatory
and the systemic proinflammatory state that is charac responses) is significantly decreased in individuals with
teristic of both groups of mice.19 Nicotine administration obesity; however, weight loss partially restores expression
significantly reduced levels of TNF in serum and abdomi- of 7nAChR.107 These findings further suggest a critical
nal adipose tissue, and decreased adipose tissue levels of role for 7nAChR in maintaining metabolic homeostasis.
TNF, IL6 and IL1 mRNAs. Nicotine also inhibited Galantamine treatment of mice with high-fat-diet-
adipose tissue expression of CCL2 and the macrophage induced obesity reduces body weight and food intake, and

NATURE REVIEWS | ENDOCRINOLOGY VOLUME 8 | DECEMBER 2012 | 749


2012 Macmillan Publishers Limited. All rights reserved
REVIEWS

selectively suppresses abdominal adiposity.20 Galantamine The importance of the vagus nerve in the regulation of
also lowers fasting blood glucose, plasma insulin and cho- metabolic and immune homeostasis and the efficacy of
lesterol levels, improves glucose homeostasis and insulin selective cholinergic modalities in alleviation of inflam-
resistance, and decreases serum alanine transaminase mation and metabolic complications indicate that specific
levels, liver weight and hepatosteatosis in these mice.20 drugs or devices that stimulate neural circuits directly to
The role of brain and peripheral cholinergic activation by activate the inflammatory reflex could be used in the
galantamine (acting as an acetylcholinesterase inhibitor treatment of obesity-related complications (Figure4).
and a positive allosteric modulator of nicotinic receptors,
including 7nAChRs)109 in mediating these beneficial Pharmacological approaches
metabolic alterations remains to be further elucidated. The efficacy of galantamine in suppressing inflammation,
Cholinergic signalling, including via 7nAChR- insulin resistance and development of fatty liver in pre-
mediated mechanisms in brain regions such as the clinical settings provides a rationale for further clinical
lateral hypothalamus, is implicated in the complex regu- studies.20 Galantamine is approved by the FDA as a treat-
lation of appetite and feeding behaviour.110 Suppression ment for Alzheimer disease, and this drug has also been
of food intake following nicotine administration is used in Europe for decades in the clinical management
thought to involve activation of 3/4nAChR in pro- of myasthenia gravis as well as other diseases. The avail-
opiomelanocortin neurons of the hypothalamic arcuate ability of data related to the previous clinical use of gal-
nucleus, which induces activation of melanocortin 4 antamine might facilitate its utilization in obesity-driven
receptors in the hypothalamic paraventricular nucleus.111 disorders, particularly in subgroups of individuals who
Thus, several cholinergic receptors, including 7nAChR, have limited capacity to benefit from lifestyle interven-
4/2nAChR and 3/4nAChR, have been implicated tions, such as elderly people. The efficacy of galantamine
in suppressing food intake and weight gain. However, in alleviating liver damage and inhibiting hepatosteatosis
7nAChR is unique in having been specifically linked to in rodents with obesity is a rationale for clinical studies to
controlling obesity-associated metabolic complications investigate the utility of this drug in the treatment of fatty
through suppression of inflammation. liver diseases (Figure4), such as nonalcoholic steato
hepatitis, which is considered to be the hepatic manifes-
Therapeutic implications tation of the metabolic syndrome. Current treatments
Obesity is directly linked with the epidemic of type2 for nonalcoholic steatohepatitis are not efficient and the
diabetes mellitus and cardiovascular disease. 112,113 disease can progress to cirrhosis and liver cancernew
Although weight loss through increased physical activ- therapies are clearly needed.117
ity and dietary alterations can be beneficial in coun- The preclinical efficacy of nAChR agonists in obesity
teracting obesity and its adverse consequences,81 these models identify nAChRs, including 7nAChR, as
lifestyle modifications are difficult to implement and can molecular targets for drug development for the treat-
be problematic to sustain. Permanent weight loss and ment of the metabolic syndrome and type2 diabetes
attenuation of dyslipidaemia and type2 diabetes mel- mellitus (Figure4). Several 7nAChR agonists, including
litus can be achieved through bariatric surgery, but these TC7020 and GTS21, have proven anti-inflammatory
operations are associated with considerable risks and properties and the latter had good tolerability in human
are currently recommended only for individuals with volunteers.118 Currently, a phaseII clinical trial of the
morbid obesity. Current pharmacological options for selective 7nAChR agonist TC6987 is ongoing in
treatment of the metabolic syndrome are limited to tar- patients with type2 diabetes mellitus.119
geting its individual components separately and, there- Obesity and the metabolic syndrome are closely
fore, multiple medications are required.81,114 Accordingly, associated with atherosclerosis and impaired cardio
an unmet need remains for new treatments for the meta- vascular regulation. Donepezil, another centrally acting
bolic syndrome and type2 diabetes mellitus, especially and FDA-approved acetylcholinesterase inhibitor, sup-
for drugs that target common steps in their pathogenesis. presses CCL2 and TNF expression, attenuates athero
The chronic inflammatory state associated with obe sclerosis and improves long-term survival following
sity is one such common step that could be targeted. chronic heart failure in rodents. 120,121 These findings
Some anti-inflammatory approaches have already been suggest that centrally acting acetylcholinesterase inhibi-
explored in the treatment of obesity-linked disorders tors could potentially be developed for the treatment
in preclinical and clinical scenarios.82,95 For example, of atherosclerosis.
patients with type2 diabetes mellitus who were treated Efferent vagus nerve-derived cholinergic activation
with a recombinant human IL1 receptor antagonist of pancreatic insulin secretion is well documented. In
(anakinra) experienced reductions in levels of IL6 and addition, human pancreatic cells secrete acetylcho-
CRP. Additionally, HbA1c levels in these patients were line, which might act through a M3 muscarinic recep-
reduced and their pancreatic cell secretory function tor-mediated mechanism as a paracrine stimulator of
improved.115 Administration of salicylatea known IKK glucose-induced -cell insulin release.122 An interesting
inhibitor in rodents, which propagates proinflammatory consideration is the hypothesis that this local acetyl
signalssignificantly improved glucose homeostasis, choline release in the pancreas could also exert a regu-
reduced free fatty acid levels and increased adiponectin latory influence on immune cells. Future studies might
levels in patients with type2 diabetes mellitus.116 test the capacity of M3 muscarinic receptor agonists to

750 | DECEMBER 2012 | VOLUME 8  www.nature.com/nrendo


2012 Macmillan Publishers Limited. All rights reserved
REVIEWS

CVD T2DM NASH

Metabolic syndrome
Disorder progression

Obesity

Target efficacy
Appetite and Glucose homeostasis Pancreatic
weight gain Inflammation Insulin resistance insulin secretion Fatty liver disease

Treatment Centrally acting 7nAChR Device-generated M3 mAChR Galantamine or other


4/2nAChR or agonists vagus nerve agonists centrally acting
3/4nAChR agonists stimulation AChE inhibitors

Figure 4 | Possible therapies based on cholinergic-based approaches for the treatment of obesity-driven disorders. Obesity
progression and the metabolic syndrome are closely associated with debilitating diseases, including T2DM, CVD and NASH.
Dysregulated immune and metabolic homeostasis is associated with inflammation underlying obesity-related disease
pathogenesis, which mediates insulin resistance and other complications. Efficacy of cholinergic agents in activating regulatory
mechanisms in the inflammatory reflex in animals indicates a rationale for developing novel treatments for humans. Cholinergic
therapeutics and devices for vagus nerve stimulation could selectively control the complex interplay between immune and
metabolic pathways. Abbreviations: CVD, cardiovascular disease; M3 mAChR, M3 muscarinic acetylcholine receptor; 7nAChR,
7 nicotinic acetylcholine receptor; 4/2nAChR, 4/2 nicotinic acetylcholine receptor; 3/4nAChR, 3/4 nicotinic
acetylcholine receptor; NASH, nonalcoholic steatohepatitis; T2DM, type 2 diabetes mellitus; VNS, vagus nerve stimulation.

promote pancreatic insulin secretion and of 7nAChR from increased physical activity, dieting or bariatric sur
agonists to suppress pancreatic inflammation in patients gery is accompanied by reductions in levels of inflamma-
with type2 diabetes mellitus (Figure 4). tory markers, amelioration of metabolic complications
Patients taking antipsychotic medication, particularly and an increase in the vagus nerve-activity component of
clozapine or olanzapine, frequently develop obesity, insu heart rate variability.15,129 A role for vagus nerve signalling
lin resistance and other metabolic complications. These in achieving and maintaining weight loss after bariatric
adverse effects represent a challenging clinical problem.123 surgery has also been reported.130 In view of these find-
Adipose tissue inflammation could contribute to the ings and the importance of the vagus nerve in immune
metabolic derangements associated with antipsychotic and metabolic regulation, restoring or augmenting vagus
treatment, as chronic administration of olanzapine to nerve activity could attenuate inflammation and other
rodents even at a dose that does not induce significant conditions associated with obesity (Figure4).
body weight gain results in enhanced adip osity and Stimulation of the vagus nerve using surgically
increased adipose tissue macrophage infiltration and TNF implanted devices is in clinical use for epilepsy and depres
expression.124 Furthermore, the adverse metabolic effects sion and, as a result, the safety profile for this approach is
of antipsychotic treatments are postulated to be related to well described.131 In patients with obesity and treatment-
their antagonism of peripheral and central M3 muscarinic resistant depression, vagus nerve stimulation is associated
receptors and their consequent suppression of positive with significant weight loss, which was positively corre-
cholinergic effects on insulin signalling and glucose lated with the degree of obesity.132 These clinical findings
homeostasis.125,126 Using galantamine treatment to alleviate are in agreement with those of several studies in obese
the metabolic syndrome in patients taking antipsychotics rodents and minipigs, in which vagus nerve stimula-
is an interesting possibility to explore. Galantamine and tion suppressed food intake and weight gain.133,134 The
other centrally-acting acetylcholinesterase inhibitors devices used in these studies predominantly stimulated
have already been tested in the context of schizophrenia afferent vagus nerve fibres; however, afferent and effer-
and shown to ameliorate selective cognitive deficits in ent vagus nerve signals converge within the dorsal vagal
patients with the disease.127 Intriguingly, progression of complex and higher brain regions, which also provide
type2 diabetes mellitus often correlates with cognitive neurophysiological circuitry to activate efferent vagus
deterioration, and new therapeutic agents for type2 dia- nerve activity that regulates metabolic homeostasis and
betes mellitus that prevent or reduce this decline in cog- immune function. Thus, stimulation of afferent and effer-
nitive function would be advantageous.128 Treatment of ent vagus nerve signalling has the potential for controlling
patients with type2 diabetes mellitus with galantamine, the inflammatory state in obesity and restoring metabolic
which has proven efficacy in ameliorating cognitive defi- regulation and insulin signalling (Figure4).
ciency, could represent a promising intervention targeting Unpublished data from our group indicate that vagus
this pathophysiologicalrelationship.127 nerve stimulation suppresses insulin resistance in
rodents. Vagus nerve activities regulating hepatic glucose
Devices for vagus nerve stimulation metabolism and cardiac function are both impaired in
Vagus nerve activity is decreased in chronic inflamma- obesity, which suggests that augmentation of efferent
tory conditions, including obesity. Weight loss resulting vagus nerve activity could be beneficial in patients with

NATURE REVIEWS | ENDOCRINOLOGY VOLUME 8 | DECEMBER 2012 | 751


2012 Macmillan Publishers Limited. All rights reserved
REVIEWS

hyperglycaemia, insulin resistance and cardiovascular of central and peripheral mechanisms in this regulation
diseases (Figure4). Vagus nerve stimulation is also being are topics of ongoing study. Additionally, intracellular
studied as a method to suppress cardiac inflammation in mechanisms by which cholinergic signals control obesity-
patients with heart failure.135 Additionally, this approach associated inflammation and modulate insulin signalling
could be studied in patients with obesity-associated dis are under investigation. 7nAChR agonists, centrally
orders, such as the metabolic syndrome and type2 dia- acting acetylcholinesterase inhibitors and direct electrical
betes mellitus. Carefully designed clinical studies are stimulation of the vagus nerve offer potential therapeutic
needed to assess the potential for treatment with selec- strategies for treating obesity, the metabolic syndrome,
tive cholinergic agonists or neurostimulating devices in type2 diabetes mellitus and other disorders associated
such patients. with obesity. The use of cholinergic modalities in com-
bination with existing or new therapeutic approaches to
Conclusions target neural, endocrine and immune functions for thera-
The inflammatory reflex mediated by the vagus nerve has peutic benefit in patients with obesity-related disorders
been successfully exploited therapeutically in preclini- should also be considered.
cal models of diseases with aetiologies characterized by
excessive inflammatory responses. Insufficient efferent Review criteria
vagus nerve cholinergic output might have a causative
Original research papers and reviews (in English) were
role in the dysfunctional immune and metabolic regu-
considered for inclusion in this manuscript on the basis
lation observed in obesity, as selective activation of the of the authors knowledge of the field and the results of
efferent cholinergic arm of the inflammatory reflex atten- PubMed and Google Scholar searches using the following
uates both inflammation and metabolic derangements. keywords: vagus, feeding behaviour, metabolism,
Although cholinergic suppression of inflammation can obesity, inflammation, metabolic syndrome,
contribute specifically to alleviating metabolic complica- type2 diabetes mellitus, and cholinergic, alone
tions, direct cholinergic effects on metabolic pathways and in combination. The authors have also reviewed the
reference lists of key manuscripts to identify additional
could also have a role in alleviating symptoms associated
relevant papers. Paper selection was limited to the past
with the metabolic syndrome and type2 diabetes mel- 35 years (19672012).
litus. These complex interactions and the contribution

1. Tracey, K.J. The inflammatory reflex. Nature 420, Obes. Relat. Metab. Disord. 20, 966969 neuroimmunomodulation. Mol. Med. 9, 125134
853859 (2002). (1996). (2003).
2. Tracey, K.J. Reflex control of immunity. Nat. Rev. 15. Karason, K., Molgaard, H., Wikstrand, J. & 24. Goehler, L.E. et al. Vagal immune-to-brain
Immunol. 9, 418428 (2009). Sjostrom, L. Heart rate variability in obesity and communication: a visceral chemosensory
3. Andersson, U. & Tracey, K.J. Reflex principles of the effect of weight loss. Am. J. Cardiol. 83, pathway. Auton. Neurosci. 85, 4959 (2000).
immunological homeostasis. Annu. Rev. 12421247 (1999). 25. Niijima, A. The afferent discharges from sensors
Immunol. 30, 313335 (2011). 16. Ziegler, D. et al. Selective contribution of for interleukin 1 in the hepatoportal system in
4. Baccala, R. et al. Sensors of the innate immune diabetes and other cardiovascular risk factors to the anesthetized rat. J. Auton. Nerv. Syst. 61,
system: their mode of action. Nat. Rev. cardiac autonomic dysfunction in the general 287291 (1996).
Rheumatol. 5, 448456 (2009). population. Exp. Clin. Endocrinol. Diabetes 114, 26. Ek, M., Kurosawa, M., Lundeberg, T. &
5. Chen, G.Y. & Nunez, G. Sterile inflammation: 153159 (2006). Ericsson,A. Activation of vagal afferents after
sensing and reacting to damage. Nat. Rev. 17. Carnethon, M.R., Jacobs, D.R. Jr, Sidney, S. & intravenous injection of interleukin-1: role of
Immunol. 10, 826837 (2010). Liu, K. Influence of autonomic nervous system endogenous prostaglandins. J. Neurosci. 18,
6. Andersson, U. & Tracey, K.J. HMGB1 is a dysfunction on the development of type 2 94719479 (1998).
therapeutic target for sterile inflammation and diabetes: the CARDIA study. Diabetes Care 26, 27. Goehler, L.E. et al. Interleukin-1 in immune
infection. Annu. Rev. Immunol. 29, 139162 30353041 (2003). cells of the abdominal vagus nerve: a link
(2011). 18. Marrero, M.B. et al. An 7 nicotinic acetylcholine between the immune and nervous systems?
7. Medzhitov, R. Inflammation 2010: new adventures receptor-selective agonist reduces weight gain J.Neurosci. 19, 27992806 (1999).
of an old flame. Cell 140, 771776 (2010). and metabolic changes in a mouse model of 28. Hansen, M.K., OConnor, K.A., Goehler, L.E.,
8. Pavlov, V.A. & Tracey, K.J. The cholinergic anti- diabetes. J. Pharmacol. Exp. Ther. 332, 173180 Watkins, L.R. & Maier, S.F. The contribution of
inflammatory pathway. Brain Behav. Immun. 19, (2010). the vagus nerve in interleukin-1-induced fever is
493499 (2005). 19. Wang, X., Yang, Z., Xue, B. & Shi, H. Activation of dependent on dose. Am. J. Physiol. Regul. Integr.
9. Tracey, K.J. Physiology and immunology of the the cholinergic antiinflammatory pathway Comp. Physiol. 280, R929R934 (2001).
cholinergic antiinflammatory pathway. J. Clin. ameliorates obesity-induced inflammation and 29. Pavlov, V.A. & Tracey, K.J. Neural regulators of
Invest. 117, 289296 (2007). insulin resistance. Endocrinology 152, 836846 innate immune responses and inflammation.
10. Pavlov, V.A. Cholinergic modulation of (2011). Cell. Mol. Life Sci. 61, 23222331 (2004).
inflammation. Int. J. Clin. Exp. Med. 1, 203212 20. Satapathy, S.K. et al. Galantamine alleviates 30. Hosoi, T., Okuma, Y., Matsuda, T. & Nomura, Y.
(2008). inflammation and other obesity-associated Novel pathway for LPS-induced afferent vagus
11. Bastard, J.P. et al. Recent advances in the complications in high-fat diet-fed mice. Mol. Med. nerve activation: possible role of nodose
relationship between obesity, inflammation, and 17, 599606 (2011). ganglion. Auton. Neurosci. 120, 104107
insulin resistance. Eur. Cytokine Netw. 17, 412 21. Borovikova, L.V. et al. Vagus nerve stimulation (2005).
(2006). attenuates the systemic inflammatory 31. de Lartigue, G., Barbier, delaSerre, C., Espero,
12. Donath, M.Y. & Shoelson, S.E. Type 2 diabetes response to endotoxin. Nature 405, 458462 E., Lee,J. & Raybould, H.E. Diet-induced obesity
as an inflammatory disease. Nat. Rev. Immunol. (2000). leads to the development of leptin resistance in
11, 98107 (2011). 22. Wang, H. et al. Nicotinic acetylcholine receptor vagal afferent neurons. Am. J. Physiol. Endocrinol.
13. Hotamisligil, G.S. Inflammation and metabolic 7 subunit is an essential regulator of Metab. 301, E187E195 (2011).
disorders. Nature 444, 860867 (2006). inflammation. Nature 421, 384388 (2003). 32. Guarini, S. et al. Adrenocorticotropin reverses
14. Richter, W.O., Geiss, H.C., Aleksic, S. & 23. Pavlov, V.A., Wang, H., Czura, C.J., hemorrhagic shock in anesthetized rats through
Schwandt, P. Cardiac autonomic nerve function Friedman,S.G. & Tracey, K.J. The cholinergic the rapid activation of a vagal anti-inflammatory
and insulin sensitivity in obese subjects. Int. J. anti-inflammatory pathway: a missing link in pathway. Cardiovasc. Res. 63, 357365 (2004).

752 | DECEMBER 2012 | VOLUME 8  www.nature.com/nrendo


2012 Macmillan Publishers Limited. All rights reserved
REVIEWS

33. Pavlov, V.A. et al. Central muscarinic cholinergic innervation in the control of energy metabolism. postprandial inflammation. Am. J. Clin. Nutr. 86,
regulation of the systemic inflammatory Biochim. Biophys. Acta. 1802, 416431 (2010). 12861292 (2007).
response during endotoxemia. Proc. Natl. Acad. 53. Owyang, C. & Heldsinger, A. Vagal control of 72. Ghanim, H. et al. Increase in plasma endotoxin
Sci. USA 103, 52195223 (2006). satiety and hormonal regulation of appetite. concentrations and the expression of Toll-like
34. Pavlov, V.A. et al. Brain acetylcholinesterase J.Neurogastroenterol. Motil. 17, 338348 (2011). receptors and suppressor of cytokine signaling-3
activity controls systemic cytokine levels through 54. Kreier, F. et al. Selective parasympathetic in mononuclear cells after a high-fat, high-
the cholinergic anti-inflammatory pathway. Brain innervation of subcutaneous and intra- carbohydrate meal: implications for insulin
Behav. Immun. 23, 4145 (2009). abdominal fatfunctional implications. J. Clin. resistance. Diabetes Care 32, 22812287
35. Lee, S.T. et al. Cholinergic anti-inflammatory Invest. 110, 12431250 (2002). (2009).
pathway in intracerebral hemorrhage. Brain Res. 55. Giordano, A. et al. White adipose tissue lacks 73. Lundman, P. et al. A high-fat meal is
1309, 164171 (2010). significant vagal innervation and accompanied by increased plasma interleukin-6
36. Pavlov, V.A. et al. Selective 7-nicotinic immunohistochemical evidence of concentrations. Nutr. Metab. Cardiovasc. Dis. 17,
acetylcholine receptor agonist GTS-21 improves parasympathetic innervation. Am. J. Physiol. Regul. 195202 (2007).
survival in murine endotoxemia and severe Integr. Comp. Physiol 291, R1243R1255 (2006). 74. Laugerette, F. et al. Emulsified lipids increase
sepsis. Crit. Care Med. 35, 11391144 (2007). 56. Wang, P.Y. et al. Upper intestinal lipids trigger a endotoxemia: possible role in early postprandial
37. Parrish, W.R. et al. Modulation of TNF release by gutbrainliver axis to regulate glucose low-grade inflammation. J. Nutr. Biochem. 22,
choline requires 7 subunit nicotinic production. Nature 452, 10121016 (2008). 5359 (2011).
acetylcholine receptor-mediated signaling. Mol. 57. Cheung, G.W., Kokorovic, A., Lam, C.K., 75. Luyer, M.D. et al. Nutritional stimulation of
Med. 14, 567574 (2008). Chari,M. & Lam, T.K. Intestinal cholecystokinin cholecystokinin receptors inhibits inflammation
38. Gallowitsch-Puerta, M. & Pavlov, V.A. Neuro- controls glucose production through a neuronal via the vagus nerve. J. Exp. Med. 202,
immune interactions via the cholinergic anti- network. Cell Metab. 10, 99109 (2009). 10231029 (2005).
inflammatory pathway. Life Sci. 80, 23252329 58. Lam, C.K. et al. Activation of N-methyl-D- 76. Bucinskaite, V., Kurosawa, M., Miyasaka, K.,
(2007). aspartate (NMDA) receptors in the dorsal vagal Funakoshi, A. & Lundeberg, T. Interleukin-1
39. Olofsson, P.S. et al. 7 nicotinic acetylcholine complex lowers glucose production. J. Biol. sensitizes the response of the gastric vagal
receptor (7nAChR) expression in bone marrow- Chem. 285, 2191321921 (2010). afferent to cholecystokinin in rat. Neurosci. Lett.
derived non-T cells is required for the 59. Nakabayashi, H., Nishizawa, M., Nakagawa, A., 229, 3336 (1997).
inflammatory reflex. Mol. Med. 18, 539543 Takeda, R. & Niijima, A. Vagal hepatopancreatic 77. Gaige, S., Abou, E., Abysique, A. & Bouvier, M.
(2012). reflex effect evoked by intraportal appearance of Effects of interactions between interleukin-1 and
40. Hamano, R. et al. Stimulation of 7 nicotinic tGLP-1. Am. J. Physiol. 271, E808E813 (1996). leptin on cat intestinal vagal mechanoreceptors.
acetylcholine receptor inhibits CD14 and the toll- 60. Balkan, B. & Li, X. Portal GLP-1 administration in J.Physiol. 555, 297310 (2004).
like receptor 4 expression in human monocytes. rats augments the insulin response to glucose 78. Doganay, M. et al. The effects of vagotomy on
Shock 26, 358364 (2006). via neuronal mechanisms. Am. J. Physiol. Regul. bacterial translocation: an experimental study.
41. Guarini, S. et al. Efferent vagal fibre stimulation Integr. Comp. Physiol. 279, R1449R1454 J.Surg. Res. 71, 166171 (1997).
blunts nuclear factor-B activation and protects (2000). 79. Eckel, R.H. Mechanisms of the components of
against hypovolemic hemorrhagic shock. 61. Vincent, K.M., Sharp, J.W. & Raybould, H.E. the metabolic syndrome that predispose to
Circulation 107, 11891194 (2003). Intestinal glucose-induced calciumcalmodulin diabetes and atherosclerotic CVD. Proc. Nutr.
42. Pavlov, V.A. & Tracey, K.J. Controlling kinase signaling in the gutbrain axis in awake Soc. 66, 8295 (2007).
inflammation: the cholinergic anti-inflammatory rats. Neurogastroenterol. Motil. 23, e282e293 80. Shoelson, S.E., Herrero, L. & Naaz, A. Obesity,
pathway. Biochem. Soc. Trans. 34, 10371040 (2011). inflammation, and insulin resistance.
(2006). 62. Obici, S., Zhang, B.B., Karkanias, G. & Gastroenterology 132, 21692180 (2007).
43. de Jonge, W.J. et al. Stimulation of the vagus Rossetti,L. Hypothalamic insulin signaling is 81. Sutherland, J.P., McKinley, B. & Eckel, R.H. The
nerve attenuates macrophage activation by required for inhibition of glucose production. Nat. metabolic syndrome and inflammation. Metab.
activating the Jak2-STAT3 signaling pathway. Nat. Med. 8, 13761382 (2002). Syndr. Relat. Disord. 2, 82104 (2004).
Immunol. 6, 844851 (2005). 63. Pocai, A. et al. Hypothalamic K(ATP) channels 82. Gregor, M.F. & Hotamisligil, G.S. Inflammatory
44. Rosas-Ballina, M. et al. Splenic nerve is required control hepatic glucose production. Nature 434, mechanisms in obesity. Annu. Rev. Immunol. 29,
for cholinergic antiinflammatory pathway control 10261031 (2005). 415445 (2010).
of TNF in endotoxemia. Proc. Natl Acad. Sci. USA 64. Pocai, A., Obici, S., Schwartz, G.J. & Rossetti, L. 83. Ouchi, N., Parker, J.L., Lugus, J.J. & Walsh, K.
105, 1100811013 (2008). A brainliver circuit regulates glucose Adipokines in inflammation and metabolic
45. Rosas-Ballina, M. et al. Acetylcholine-synthesizing homeostasis. Cell Metab. 1, 5361 (2005). disease. Nat. Rev. Immunol. 11, 8597 (2011).
T cells relay neural signals in a vagus nerve 65. Shimazu, T., Matsushita, H. & Ishikawa, K. 84. Tilg, H. & Moschen, A.R. Adipocytokines:
circuit. Science 334, 98101 (2011). Cholinergic stimulation of the rat hypothalamus: mediators linking adipose tissue, inflammation
46. Ghia, J.E., Blennerhassett, P., Kumar- effects of liver glycogen synthesis. Science 194, and immunity. Nat. Rev. Immunol. 6, 772783
Ondiveeran, H., Verdu, E.F. & Collins, S.M. 535536 (1976). (2006).
The vagus nerve: a tonic inhibitory influence 66. Matsushita, H., Ishikawa, K. & Shimazu, T. 85. Olefsky, J.M. & Glass, C.K. Macrophages,
associated with inflammatory bowel disease Chemical coding of the hypothalamic neurones inflammation, and insulin resistance. Annu. Rev.
in a murine model. Gastroenterology 131, in metabolic control. I. Acetylcholine-sensitive Physiol. 72, 219246 (2010).
11221130 (2006). neurones and glycogen synthesis in liver. Brain 86. Nishimura, S. et al. CD8+ effector T cells
47. Bonaz, B. The cholinergic anti-inflammatory Res. 163, 253261 (1979). contribute to macrophage recruitment and
pathway and the gastrointestinal tract. 67. Ruiz, d.A.I, Gautam, D., Guettier, J.M. & adipose tissue inflammation in obesity. Nat.
Gastroenterology 133, 13701373 (2007). Wess,J. Novel insights into the function of -cell Med. 15, 914920 (2009).
48. Leib, C. et al. Role of the cholinergic M3 muscarinic acetylcholine receptors: 87. Nathan, C. Epidemic inflammation: pondering
antiinflammatory pathway in murine autoimmune therapeutic implications. Trends Endocrinol. obesity. Mol. Med. 14, 485492 (2008).
myocarditis. Circ. Res. 109, 130140 (2011). Metab. 22, 7480 (2011). 88. Baker, R.G., Hayden, M.S. & Ghosh, S. NF-B,
49. van Maanen, M.A., Vervoordeldonk, M.J. & 68. Kampe, J., Tschop, M.H., Hollis, J.H. & inflammation, and metabolic disease. Cell
Tak,P.P. The cholinergic anti-inflammatory Oldfield,B.J. An anatomic basis for the Metab. 13, 1122 (2011).
pathway: towards innovative treatment of communication of hypothalamic, cortical and 89. Vandanmagsar, B. et al. The NLRP3
rheumatoid arthritis. Nat. Rev. Rheumatol. 5, mesolimbic circuitry in the regulation of energy inflammasome instigates obesity-induced
229232 (2009). balance. Eur. J. Neurosci. 30, 415430 (2009). inflammation and insulin resistance. Nat. Med.
50. Koopman, F.A. et al. Restoring the balance of 69. Morton, G.J., Cummings, D.E., Baskin, D.G., 17, 179188 (2011).
the autonomic nervous system as an innovative Barsh, G.S. & Schwartz, M.W. Central nervous 90. Masters, S.L. et al. Activation of the NLRP3
approach to the treatment of rheumatoid system control of food intake and body weight. inflammasome by islet amyloid polypeptide
arthritis. Mol. Med. 17, 937948 (2011). Nature 443, 289295 (2006). provides a mechanism for enhanced IL-1 in
51. Huston, J.M. & Tracey, K.J. The pulse of 70. Saper, C.B., Chou, T.C. & Elmquist, J.K. The type 2 diabetes. Nat. Immunol. 11, 897904
inflammation: heart rate variability, the cholinergic need to feed: homeostatic and hedonic control (2010).
anti-inflammatory pathway and implications for of eating. Neuron 36, 199211 (2002). 91. Musso, G., Gambino, R. & Cassader, M.
therapy. J. Intern. Med. 269, 4553 (2011). 71. Erridge, C., Attina, T., Spickett, C.M. & Interactions between gut microbiota and host
52. Yi, C.X., la Fleur, S.E., Fliers, E. & Kalsbeek, A. Webb,D.J. A high-fat meal induces low-grade metabolism predisposing to obesity and
The role of the autonomic nervous liver endotoxemia: evidence of a novel mechanism of diabetes. Annu. Rev. Med. 62, 361380 (2011).

NATURE REVIEWS | ENDOCRINOLOGY VOLUME 8 | DECEMBER 2012 | 753


2012 Macmillan Publishers Limited. All rights reserved
REVIEWS

92. Delzenne, N.M., Neyrinck, A.M., Backhed, F. & 110. Jo, Y.H., Wiedl, D. & Role, L.W. Cholinergic type 2 diabetes mellitus in the elderly. Nat. Rev.
Cani, P.D. Targeting gut microbiota in obesity: modulation of appetite-related synapses in Endocrinol. 7, 108114 (2011).
effects of prebiotics and probiotics. Nat. Rev. mouse lateral hypothalamic slice. J. Neurosci. 129. Maser, R.E., Lenhard, M.J., Irgau, I. &
Endocrinol. 7, 639646 (2011). 25, 1113311144 (2005). Wynn,G.M. Impact of surgically induced weight
93. Cani, P.D. et al. Metabolic endotoxemia initiates 111. Mineur, Y.S. et al. Nicotine decreases food loss on cardiovascular autonomic function: one-
obesity and insulin resistance. Diabetes 56, intake through activation of POMC neurons. year follow-up. Obesity (Silver Spring) 15,
17611772 (2007). Science 332, 13301332 (2011). 364369 (2007).
94. Tilg, H. The role of cytokines in non-alcoholic fatty 112. Grundy, S.M. Metabolic syndrome pandemic. 130. Bueter, M. et al. Vagal sparing surgical technique
liver disease. Dig. Dis. 28, 179185 (2010). Arterioscler. Thromb. Vasc. Biol. 28, 629636 but not stoma size affects body weight loss in
95. Tilg, H. & Moschen, A.R. Inflammatory (2008). rodent model of gastric bypass. Obes. Surg. 20,
mechanisms in the regulation of insulin 113. James, W.P. The epidemiology of obesity: the 616622 (2010).
resistance. Mol. Med. 14, 222231 (2008). size of the problem. J. Intern. Med. 263, 131. Clancy, J.A., Deuchars, S.A. & Deuchars, J.
96. Ridker, P. M. Clinical application of C-reactive 336352 (2008). 2012 GL Brown Lecture The Wonders of the
protein for cardiovascular disease detection and 114. Vetter, M.L., Faulconbridge, L.F., Webb, V.L. & Wanderer. Exp. Physiol. http://dx.doi.org/
prevention. Circulation 107, 363369 (2003). Wadden, T.A. Behavioral and pharmacologic 10.1113/expphysiol.2012.064543.
97. Scherer, T. et al. Brain insulin controls adipose therapies for obesity. Nat. Rev. Endocrinol. 6, 132. Pardo, J.V. et al. Weight loss during chronic,
tissue lipolysis and lipogenesis. Cell Metab. 13, 578588 (2010). cervical vagus nerve stimulation in depressed
183194 (2011). 115. Larsen, C.M. et al. Interleukin-1-receptor patients with obesity: an observation. Int. J.
98. Howard, J.K. & Flier, J.S. Attenuation of leptin antagonist in type 2 diabetes mellitus. N. Engl. J. Obes. (Lond.) 31, 17561759 (2007).
and insulin signaling by SOCS proteins. Trends Med. 356, 15171526 (2007). 133. Bugajski, A.J. et al. Effect of long-term vagal
Endocrinol. Metab. 17, 365371 (2006). 116. Goldfine, A.B., Fonseca, V. & Shoelson, S.E. stimulation on food intake and body weight
99. de La Serre, C.B. et al. Propensity to high-fat Therapeutic approaches to target inflammation in during diet induced obesity in rats. J. Physiol.
diet-induced obesity in rats is associated with type 2 diabetes. Clin. Chem. 57, 162167 (2011). Pharmacol. 58 (Suppl. 1), 512 (2007).
changes in the gut microbiota and gut 117. Cohen, J.C., Horton, J.D. & Hobbs, H.H. Human 134. Val-Laillet, D., Biraben, A., Randuineau, G. &
inflammation. Am. J. Physiol. Gastrointest. Liver fatty liver disease: old questions and new Malbert, C.H. Chronic vagus nerve stimulation
Physiol. 299, G440G448 (2010). insights. Science 332, 15191523 (2011). decreased weight gain, food consumption and
100. Lee, J. et al. Glucose-sensing by gut endocrine 118. Kitagawa, H. et al. Safety, pharmacokinetics, and sweet craving in adult obese minipigs. Appetite
cells and activation of the vagal afferent pathway effects on cognitive function of multiple doses of 55, 245252 (2010).
is impaired in a rodent model of type 2 diabetes GTS-21 in healthy, male volunteers. 135. Schwartz, P.J. et al. Long term vagal stimulation
mellitus. Am. J. Physiol. Regul. Integr. Comp. Neuropsychopharmacology 28, 542551 (2003). in patients with advanced heart failure: first
Physiol. 302, R657R666 (2011). 119. US National Library of Medicine. ClinicalTrials.gov experience in man. Eur. J. Heart Fail. 10,
101. Lam, T.K. et al. Hypothalamic sensing of [online], http://clinicaltrials.gov/ct2/show/NCT0 884891 (2008).
circulating fatty acids is required for glucose 1293669?term=Targacept&rank=11 (2012). 136. Friedman, J.M. & Halaas, J.L. Leptin and the
homeostasis. Nat. Med. 11, 320327 (2005). 120. Inanaga, K. et al. Acetylcholinesterase inhibitors regulation of body weight in mammals. Nature
102. Morgan, K., Obici, S. & Rossetti, L. Hypothalamic attenuate atherogenesis in apolipoprotein 395, 763770 (1998).
responses to long-chain fatty acids are E-knockout mice. Atherosclerosis 213, 5258 137. La Cava, A. & Matarese, G. The weight of leptin in
nutritionally regulated. J. Biol. Chem. 279, (2010). immunity. Nat. Rev. Immunol. 4, 371379 (2004).
3113931148 (2004). 121. Handa, T. et al. Anti-Alzheimers drug, donepezil, 138. Maedler, K. et al. Leptin modulates cell
103. Ribeiro, R.T., Lautt, W.W., Legare, D.J. & markedly improves long-term survival after expression of IL-1 receptor antagonist and
Macedo, M.P. Insulin resistance induced by chronic heart failure in mice. J. Card. Fail. 15, release of IL-1 in human islets. Proc. Natl Acad.
sucrose feeding in rats is due to an impairment 805811 (2009). Sci. USA 101, 81388143 (2004).
of the hepatic parasympathetic nerves. 122. Rodriguez-Diaz, R. et al. cells secrete 139. Hotamisligil, G.S., Arner, P., Caro, J.F.,
Diabetologia 48, 976983 (2005). acetylcholine as a non-neuronal paracrine signal Atkinson,R.L. & Spiegelman, B.M. Increased
104. Miller, A.W., Sims, J.J., Canavan, A., Hsu, T. & priming cell function in humans. Nat. Med. 17, adipose tissue expression of tumor necrosis
Ujhelyi, M.R. Impaired vagal reflex activity in 888892 (2011). factor- in human obesity and insulin resistance. J.
insulin-resistant rats. J. Cardiovasc. Pharmacol. 123. Citrome, L. & Volavka, J. Consensus development Clin. Invest. 95, 24092415 (1995).
33, 698702 (1999). conference on antipsychotic drugs and obesity 140. Leinonen, E. et al. Insulin resistance and
105. Gautam, D. et al. A critical role for cell M3 and diabetes: response to consensus statement. adiposity correlate with acute-phase reaction
muscarinic acetylcholine receptors in regulating J. Clin. Psychiatry 66, 10731074 (2005). and soluble cell adhesion molecules in type 2
insulin release and blood glucose homeostasis 124. Victoriano, M. et al. Olanzapine-induced diabetes. Atherosclerosis 166, 387394 (2003).
in vivo. Cell Metab. 3, 449461 (2006). accumulation of adipose tissue is associated 141. Kanda, H. et al. MCP-1 contributes to
106. Liu, R.H., Mizuta, M. & Matsukura, S. The with an inflammatory state. Brain Res. 1350, macrophage infiltration into adipose tissue,
expression and functional role of nicotinic 167175 (2010). insulin resistance, and hepatic steatosis in
acetylcholine receptors in rat adipocytes. 125. Teff, K.L. & Kim, S.F. Atypical antipsychotics obesity. J. Clin. Invest. 116, 14941505 (2006).
J.Pharmacol. Exp. Ther. 310, 5258 (2004). and the neural regulation of food intake and 142. Steppan, C.M. et al. The hormone resistin links
107. Cancello, R. et al. The nicotinic acetylcholine peripheral metabolism. Physiol. Behav. 104, obesity to diabetes. Nature 409, 307312
receptor 7 in subcutaneous mature adipocytes: 590598 (2011). (2001).
downregulation in human obesity and modulation 126. Weston-Green, K., Huang, X.F., Lian, J. & Deng,C.
by diet-induced weight loss. Int. J. Obes. (Lond.) Effects of olanzapine on muscarinic M3 receptor Acknowledgements
http://dx.doi.org/10.1038/ijo.2011.275. binding density in the brain relates to weight gain, The authors research work is supported in part by
108. Waldburger, J.M. et al. Spinal p38 MAP kinase plasma insulin and metabolic hormone levels. Eur. grants from the National Institute of General Medical
regulates peripheral cholinergic outflow. Arthritis Neuropsychopharmacol. 22, 364373 (2011). Sciences, NIH. The authors thank Peder S. Olofsson
Rheum. 58, 29192921 (2008). 127. Ribeiz, S.R. et al. Cholinesterase inhibitors as for critical reading of the manuscript.
109. Albuquerque, E.X., Santos, M.D., Alkondon, M., adjunctive therapy in patients with schizophrenia
Pereira, E.F. & Maelicke, A. Modulation of and schizoaffective disorder: a review and meta- Author contributions
nicotinic receptor activity in the central nervous analysis of the literature. CNS Drugs 24, Both authors contributed equally to researching data
system: a novel approach to the treatment of 303317 (2010). for the article, writing the manuscript, discussions of
Alzheimer disease. Alzheimer Dis. Assoc. Disord. 128. Strachan, M.W., Reynolds, R.M., Marioni, R.E. the content, and review or editing of the manuscript
15 (Suppl. 1), S19S25 (2001). & Price, J.F. Cognitive function, dementia and before submission.

754 | DECEMBER 2012 | VOLUME 8  www.nature.com/nrendo


2012 Macmillan Publishers Limited. All rights reserved

Potrebbero piacerti anche