ArticlePDF Available

The "Big Bang" in obese fat: Events initiating obesity-induced adipose tissue inflammation

Authors:

Abstract

Obesity is associated with the accumulation of pro-inflammatory cells in visceral adipose tissue (VAT), which is an important underlying cause of insulin resistance and progression to diabetes mellitus type 2 (DM2). Whereas the role of pro-inflammatory cytokines in disease development is established, the initiating events leading to immune cell activation remain elusive. Lean adipose tissue is predominantly populated with regulatory cells, such as eosinophils and type 2 innate lymphocytes. These cells maintain tissue homeostasis through the excretion of type 2 cytokines, such as IL-4, IL-5 and IL-13, which keep adipose tissue macrophages (ATMs) in an anti-inflammatory, M2-like state. Diet-induced obesity is associated with the loss of tissue homeostasis and development of type 1 inflammatory responses in VAT, characterized by IFN-γ. A key event is a shift of ATMs towards an M1 phenotype. Recent studies show that obesity-induced adipocyte hypertrophy results in upregulated surface expression of stress markers. Adipose stress is detected by local sentinels, such as NK cells and CD8(+) T cells, which produce IFN-γ, driving M1 ATM polarization. A rapid accumulation of pro-inflammatory cells in VAT follows, leading to inflammation. In this review, we provide an overview of events leading to adipose tissue inflammation, with a special focus on adipose homeostasis and the obesity-induced loss of homeostasis which marks the initiation of VAT-inflammation. This article is protected by copyright. All rights reserved. This article is protected by copyright. All rights reserved.
Received: 03-Apr-2015; Revised: 07-Jul-2015; Accepted: 22-Jul-2015
This article has been accepted for publication and undergone full peer review but has not been through the copyediting,
typesetting, pagination and proofreading process, which may lead to differences between this version and the Version of
Record. Please cite this article as doi: 10.1002/eji.201545502.
This article is protected by copyright. All rights reserved. 1
The “Big Bang in obese fat:
events initiating obesity-induced adipose tissue inflammation
Felix M. Wensveen1,21,41,4, Tamara Turk Wensveen31*
1Department of Histology & Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
2Department of Experimental Immunology, Amsterdam Medical Centre, Amsterdam, The
Netherlands
3Department of Internal Medicine, University Hospital Rijeka, Rijeka, Croatia
4These authors contributed equally
*Correspondence to: bojan.polic@medri.uniri.hr, tel. +385 51 651 171, fax. +385 51 651 176
Abstract
Obesity is associated with the accumulation of pro-inflammatory cells in visceral adipose
tissue (VAT), which is an important underlying cause of insulin resistance and progression to
diabetes mellitus type 2 (DM2). Whereas the role of pro-inflammatory cytokines in disease
development is established, the initiating events leading to immune cell activation remain elusive.
Lean adipose tissue is predominantly populated with regulatory cells, such as eosinophils and type 2
innate lymphocytes. These cells maintain tissue homeostasis through the excretion of type 2
cytokines, such as IL-4, IL-5 and IL-13, which keep adipose tissue macrophages (ATMs) in an anti-
inflammatory, M2-like state. Diet-induced obesity is associated with the loss of tissue homeostasis
and development of type 1 inflammatory responses in VAT, characterized by IFN-
shift of ATMs towards an M1 phenotype. Recent studies show that obesity-induced adipocyte
hypertrophy results in upregulated surface expression of stress markers. Adipose stress is detected
by local sentinels, such as NK cells and CD8+ T cells, which produce IFN-, driving M1 ATM
polarization. A rapid accumulation of pro-inflammatory cells in VAT follows, leading to inflammation.
In this review, we provide an overview of events leading to adipose tissue inflammation, with a
special focus on adipose homeostasis and the obesity-induced loss of homeostasis which marks the
initiation of VAT-inflammation.
This article is protected by copyright. All rights reserved. 2
Key Words: Obesity, inflammation, adipose tissue, IFN-, TNF, adiponectin, NK cells, macrophages,
insulin resistance, diabetes mellitus type 2
Introduction
Over the past few decades, we have seen a dramatic worldwide increase in the incidence of
obesity and its associated pathologies, such as insulin resistance, which contributes to the
development of metabolic syndrome and diabetes mellitus type 2 (DM2)[1]. Metabolic syndrome is
a cluster of conditions such as elevated blood glucose, elevated blood pressure, excess body fat and
abnormal lipid (cholesterol) levels, which together increase the risk of diabetes mellitus and
cardiovascular disease. DM2 is characterized by high blood glucose levels, which is a direct result of
reduced systemic sensitivity to the anabolic hormone insulin. An important underlying cause of
obesity-induced insulin resistance is chronic low-grade systemic inflammation (reviewed in [2]). The
long-term presence of pro-inflammatory cytokines in the blood blunts the signal transduction
capacity of the insulin receptor in insulin-sensitive tissues [3]. Obesity-induced systemic
inflammation is thought to originate predominantly in adipose tissue. The human body contains
various types of fat depots, generally divided in white and brown fat. The role of brown adipose
tissue is to produce body heat and in humans is mostly found in newborns, even though adults do
have small amounts of this type of fat [4]. The role of white adipose tissue (WAT) is to store
nutrients in the form of a single large fat droplet. In addition, WAT is an important sensor of the
metabolic state of an organism and is therefore one of the main endocrine organs in the body [5].
WAT can be found at multiple sites in the body and can be further subdivided based on location and
on differences in precursor cells that give rise to these organs [6]. For example, retroperitoneal WAT
is derived from a precursor that requires expression of the transcription factors Myf5 and Pax3,
whereas these genes are not essential for the development of mesenteric WAT. In contrast, male
perigonadal WAT does not require Myf5, but is partially dependent on Pax3 for its development in
mice [6].
Of these various fat tissues, the intra-abdominal fat depots, collectively referred to as
visceral adipose tissue (VAT), have been shown to be the predominant source of chronic systemic
inflammation and most important for the development of DM2 [2]. When the perigonadal fat pads
of mice were surgically removed two weeks before initiation of HFD feeding, a significant reduction
of glucose intolerance and insulin resistance was observed in mouse models of obesity-induced DM2
This article is protected by copyright. All rights reserved. 3
[7]. VAT contains a relatively large population of immune cells, which changes dramatically in its
composition during the development of obesity [2]. Recently, various studies have provided further
insight in the earliest changes that occur within the immune cell composition of VAT in response to
diet-induced obesity (DIO). In this review, we provide a brief overview of the events that ultimately
result in adipose tissue inflammation and systemic insulin resistance. We focus on the initial stages
of immune cell activation, which represent the equivalent of the astronomical Bfor adipose
tissue inflammation. Whereas many of these events will (partially) overlap, we present them as
consecutive events to facilitate understanding.
Adipose tissue in homeostasis
The immune system plays an important role in the control of adipose tissue structure and
homeostasis. Two main functions of the immune system in fat can be distinguished: (i) inhibition of
tissue inflammation and (ii) tissue (re)modeling. Much attention has been given to regulation of
immune cell activation in VAT, since loss of immune cell inhibitory mechanisms drives the tissue
inflammation that contributes to insulin resistance. However, various immunological deficiencies
(described below) result either in reduced or increased adipose tissue mass, with obvious
implications for the endocrine function of fat. Therefore, a better understanding of tissue
remodeling by the immune system may have important implications for the current pandemic of
obesity.
Inhibition of immune cell activation in adipose tissue
Under lean conditions, adipose tissue is populated by a number of immune cells (Figure 1).
These cells either inhibit immune cell activation or promote a Th2-type response, characterized by
the production of cytokines such as IL-4, IL-5 and IL-13 [8]. In lean tissue, macrophages have been
shown to be a dominant immune cell population, and the majority of these cells have an M2-like (or
alternatively activated) phenotype (Figure 1) [9]. Lean adipose tissue macrophages (ATMs) express
arginase-1, which inhibits iNOS activity, and produce anti-inflammatory molecules, such as IL-10 and
IL-1Ra [9]. Importantly, under non-obese conditions, macrophages have been shown to play a key
role in inhibiting immune cell activation in murine fat. In mouse models where ATMs fail to respond
to M2-polarizing stimuli, an increase of pro-inflammatory cytokines, such as TNF and IL-, has been
This article is protected by copyright. All rights reserved. 4
observed. [10, 11]. The M2 phenotype of ATMs is maintained by a number of immune cells, as well
as by adipocytes. IL-4 has been shown to be an important cytokine that drives M2 polarization [10].
In adipose tissue, eosinophils are the dominant source of IL-4. Deficiency of these cells in dblGATA
mice, or hyper-eosinophilia in IL-5 transgenic animals, has been shown to result in reduced or
increased numbers of M2-like macrophages respectively [11]. VAT-resident eosinophils depend on
IL-5 for their survival, which is mainly produced by type 2 innate lymphoid cells (ILC2s) in this organ
[12]. Elimination of any component of the ILC2/eosinophil/M2-ATM axis has been shown to result in
an increase in pro-inflammatory cytokines in VAT and increased sensitivity to obesity-induced
development of insulin resistance, making this particular axis one of the dominant regulatory
mechanisms of adipose tissue homeostasis [11-13].
Invariant-chain natural killer T (iNKT) cells represent a second immune cell population which
sustains M2 ATMs in VAT (Figure 1). These cells are present at relatively high frequency in lean fat
and recognize lipid antigens in the context of CD1d [14]. Lack of iNKT cells as a result of  
CD1d deficiency in mice leads to reduced adipose tissue levels of IL-4 and IL-13, as well as to
increased pro-inflammatory ATM numbers (reviewed in [15]). The role of iNKT cells may change in
response to obesity, as it has been reported that these cells can also promote insulin resistance
following DIO [15]. The exact mechanism via which iNKT cells inhibit adipose tissue inflammation
under homeostasis, yet promote it in models of DIO, has yet to be resolved.
Regulatory T (Treg) cells represent a second T-cell subset that is directly involved in the
inhibition of adipose tissue inflammation. CD4+ T cells are the most predominant T cells in adipose
tissue and compared to other tissues, a very large fraction of these is Foxp3positive, which was
shown to depend on the cytokine IL-33 [16] [17]. Regulatory T cells
in VAT repress immune cell activation through production of the anti-inflammatory cytokine IL-10
[18]. Experimental ablation of Treg cells by injection of diphtheria toxin in mice expressing the
diphtheria toxin receptor under the Foxp3 promoter was shown to result in an increase of pro-
inflammatory cytokines such as TNF, IL-6 and RANTES in fat [18]. Importantly, elimination of Treg
cells acutely reduced insulin sensitivity in these animals [18], whereas transfer of Treg cells into T-
cell-deficient animals improved insulin sensitivity upon DIO [19].
In addition to immune cells, adipocytes and adipose stroma contribute to tissue
homeostasis. Adipose tissue excretes a number of factors, generally referred to as adipokines,
which play an important role in the regulation of systemic metabolism [20]. Many of these factors
share homologies with cytokines and have profound impact on immune cell behavior. One of the
This article is protected by copyright. All rights reserved. 5
most well characterized adipokines is adiponectin (Figure 1). This molecule shares functional
homology with insulin, and has been shown to impair gluconeogenesis in the liver and promote
glucose uptake [21]. In addition, adiponectin has a strong anti-inflammatory effect [22]. In vitro
stimulation assays showed that the adiponectin receptor is expressed at relatively high levels on M2
macrophages, whereas M1 polarization results in its downregulation, explaining the stronger effect
of adiponectin on the former ATM subset [22]. Adiponectin inhibits NF-B activation and promotes
IL-10 and IL-1Ra production by macrophages. Moreover, adiponectin suppresses TLR4 signaling [22],
which has been shown to be important for diet-induced insulin resistance [23].
Immunological control of adipose tissue structure
Various immune cell subsets have been implicated in the control of adipose tissue
remodeling. KitW-sh mice, which lack mature mast cells, have been shown to have strikingly less
adipose tissue mass than wild type animals [24]. In addition, KitW-sh mice show reduced adipose
tissue expansion upon feeding with a high-fat diet, when compared with that in wild type controls
[24]. Mast cells were shown to promote micro-vessel growth by excreting IL-6 and IFN- 
appear to be essential for healthy adipose tissue formation and expansion [24]. In contrast, mice
deficient for IL-17 demonstrated increased adipose tissue mass gain in response to HFD [25]. A
normal, low-fat diet did not result in altered adipose tissue mass in these animals, suggesting that
adipogenesis induced by the abundant presence of nutrients differs from basic ontogenesis of
adipose organs. Immune cell-derived stimuli are also capable of inhibiting adipogenesis. IL-5
transgenic animals, which suffer from eosinophilia, have been shown to have reduced adipose tissue
mass, whereas  mice, which lack eosinophils, have larger fat pads than wild type animals,
both under lean and obese conditions [11]. Deletion of iNKT cells, as a result of genetic deficiency for
an increase of adipose tissue mass only after high-fat feeding [26, 27]. The
mechanisms via which these cells control white adipose tissue proliferation is currently unclear.
A cytokine of particular interest for adipose tissue homeostasis, because it affects both
immune cell behavior and adipose tissue remodeling, is IL-33 [28]. IL-33 is a member of the IL-1
superfamily and binds to the receptor ST2, which is highly expressed on mast cells, Th2 CD4+ T cells
and ILC2s [29]. Under homeostatic conditions IL-33 is mainly expressed by epithelial cells and tissue
stroma [30]. Upon infection, IL-33 expression is highly induced in many tissues and it was therefore
originally classified as a pro-inflammatory mediator which drives anti-helminth immune responses
[29]. Under non-inflammatory conditions, IL-33 sustains type 2 immune cells, including ILC2s and M2
macrophages, in order to maintain tissue homeostasis (reviewed in [29]). In VAT, IL-33 is abundantly
This article is protected by copyright. All rights reserved. 6
expressed in adipose tissue stroma, predominantly by endothelial cells and fibroblast-like reticular
cells [29, 30]. Adipose tissue Treg cells express high levels of ST2 and deficiency of this receptor
results in a specific loss of Treg cells in fat, but not in other organs [16]. IL-33 also plays a role in the
maintenance of adipose tissue eosinophils, as blocking of ST2 was shown to result in a significant
reduction in the number of these cells, independently of ILC2s [31].
Of particular interest is the effect of IL-33 on ILC2s. Exogenous administration of IL-33
increases the number of ILC2s in VAT, with a concomitant increase of IL-5 levels and eosinophil cell
numbers [12]. Strikingly, in response to IL-33, ILC2s regulate adipocyte phenotype and function. For
example, IL-33 deficiency results in increased body mass and increased formation of glucose
intolerance in response to high-fat feeding, due to a lack of Type 2 ILCs [16, 32] 
(Brown in white) cell numbers, which are UCP1-expressing adipocytes in white fat with a brown
adipocyte phenotype, were shown to be decreased in these animals [32, 33]. Two recent studies
have shown that IL-33 activates ILC2s to produce IL-13 and the endogenous opioid Met-enkephalin,
which drives adipocyte precursors to differentiate into Brite cells
[32, 33]. Future studies are required to show whether other cells that produce IL-13 in fat, such as
iNKT cells, are also capable of skewing pre-adipocyte differentiation. Nevertheless, since the role of
IL-33 in beiging is most prominent in subcutaneous fat, it comprises most likely a different biological
mechanism than its anti-diabetic and anti-adipogenic effects on VAT. In addition, the biological
significance of beiging requires further study, as it is currently unclear whether it protects against
development of insulin resistance following DIO.
Reaching critical mass: obesity-driven immune cell accumulation in VAT
The early phases of DIO in visceral adipose fat are characterized by an increase in the
amount of fat per adipocyte and by an accumulation of immune cells which initially are of limited
inflammatory capacity. The most commonly used animal model for the induction of obesity and
obesity-induced insulin resistance is by feeding animals, usually mice, with a high-fat diet (HFD).
Within the first weeks after start of HFD feeding, mice accumulate neutrophils, macrophages and NK
cells in VAT. Data showing the biological relevance of these cells in VAT will be discussed below.
This article is protected by copyright. All rights reserved. 7
Neutrophils
Already in the first days after initiation of HFD feeding in mice, neutrophil numbers rapidly increase
in adipose tissue and produce the proteolytic enzyme elastase [34]. Even after long term (>3
months) HFD feeding, however, neutrophils remain a minor fraction of adipose tissue leukocytes.
Nevertheless, elastase deficiency or chemical inhibition of elastase was shown to result in enhanced
insulin sensitivity compared to wild type mice upon 12 weeks of HFD feeding. In vivo administration
of exogenous elastase resulted in acute reduction of insulin sensitivity in hepatocytes [34]. These
experiments demonstrate the importance of elastase in metabolic disease, even though it is
currently unclear how this enzyme contributes to adipose tissue inflammation. The immediate
trigger for neutrophil infiltration into the VAT following DIO is also unknown. It has been shown in
humans that acute lipid overload induces an inflammatory boost, as demonstrated by an increase of
circulating MCP1 and C-reactive protein [35]. Alternatively, HFD is thought to induce acute changes
in the adipose tissue micro-environment, such as alterations in oxygen consumption, following a
brief surge in adipocyte precursor proliferation [36]. This acute stress may be involved in recruiting
neutrophils to this site in the first days following initiation of high-fat feeding.
Macrophages and NK cells
Compared with that of neutrophils, the increase in the numbers of adipose tissue
macrophages and NK cells appears to be delayed, occurring in weeks, rather than days after the start
of HFD feeding. Whereas neutrophils in VAT are thought to be primarily of peripheral origin [34],
ATMs and NK cells appear to increase partially or completely through proliferation of tissue resident
populations [7, 37, 38]. When mice fed a HFD were intravenously injected with labeled NK cells,
tracing of the labeled cells demonstrated that only a small number of NK cells reach the adipose
tissues from the periphery. In contrast, BrdU labeling, as a marker of proliferation, was significantly
increased in adipose-tissue resident NK cells, but not in splenic NK cells following HFD [7]. This
suggests that the increase in NK cells in VAT is due to proliferation of the tissue-resident population
rather than influx from the periphery. The origin of increased macrophage populations in the VAT in
response to HFD appears to be more complex. One study demonstrates that local proliferation of
these cells is the dominant source of cellular increase [38]. Other studies, in which the capacity of
macrophages to respond to chemo-attractants was blocked, showed reduced macrophage cell
numbers in VAT due to decreased peripheral influx [37, 39]. Possibly, both depots contribute to the
ATM pool in the case of chronic inflammation in VAT.
This article is protected by copyright. All rights reserved. 8
Regulatory cells: ILC2s, Treg cells and eosinophils
In addition to the accumulation of pro-inflammatory cells in VAT, DIO is also associated with
a decrease in anti-inflammatory immune cells. In humans and mice, ILC2s were shown to be
reduced in adipose tissue of obese individuals, both relative to the total number of leukocytes and
per gram of adipose tissue [32]. This decrease appears to involve the inhibition of ILC2s to respond
to IL-33 in the presence of the type 1 cytokine IFN-[40]. Similarly, Treg cells decrease in number per
gram of fat following DIO, and especially relative to the number of effector CD8+ and CD4+ T cells.
The relative inhibitory capacity of this cell population therefore decreases in response to DIO, which
was shown to be an important contributor to VAT inflammation [16, 18, 19]. Finally, eosinophils also
decrease in number following DIO, possibly as a result of the decreased numbers of ILC2s, which
produce the IL-5 required for the maintenance of this population [11, 12]. Thus, the regulatory
capacity of inhibitory immune cell subsets in VAT decreases due to a relative decrease in the
contribution of these cells per gram of fat and as a percentage of the overall immune cell pool.
Adipose tissue
The crucial trigger for the increase of immune cells in adipose tissue in response to DIO is still
unclear, but is likely to be derived from adipocytes. Via lineage tracing of adipocyte precursors, it
was shown that the increase in adipose tissue mass early after HFD feeding is predominantly the
result from adipocyte hypertrophy [36, 41]. Despite an early surge in pre-adipocyte proliferation,
only prolonged exposure to HFD (> 4 weeks) increases mature adipocyte cell numbers (hyperplasia)
[36, 41]. With the increase of adipose tissue mass, several changes occur in adipokine production.
Most notably, adiponectin production drops, which results in decreased glucose uptake and reduced
anti-inflammatory regulation of the local tissue environment [22]. Instead, adipocytes start
producing more leptin. Leptin affects the hypothalamus, where it triggers satiety signals, thereby
directly inhibiting the effects of ghrelin, a hormone produced in the gut that stimulates hunger [22].
Leptin levels in the blood closely correlate with the amount of adipose tissue mass, both in humans
and mice [42]. In addition to its central effects, leptin has a profound effect on the immune system.
The leptin receptor, also known as LEP-R or CD295, is expressed on most immune cells including
neutrophils, macrophages and NK cells. LEP-R shares structural homology with the IL-6 receptor and
also signals through Stat3 [43]. Mice that are deficient for leptin (ob/ob) or for LEP-R (db/db) are
obese due to increased food intake as a result of never feeling sated. In addition, they have a strong
This article is protected by copyright. All rights reserved. 9
reduction in functional immune cells, such as (regulatory) T cells [44], NK cells and dendritic cells
(reviewed in [45]). Importantly, T cells and B cells have been shown to increase expression of LEP-R
in response to activation [46]. Moreover, survival of activated lymphocytes was enhanced when
leptin was added to the cell culture [46]. Increased leptin production by adipose tissue may be an
important initial trigger for immune cell increase in VAT in response to obesity. Indeed, in the first
weeks after start of HFD feeding, macrophage and NK-cell numbers and functionality are specifically
increased in VAT, whereas they are reduced in ob/ob mice [7, 47]. Moreover, leptin has been shown
to promote NK-cell survival in the bone marrow [48] and intravenous administration of leptin
resulted in a specific increase of granulocytes, monocytes and NK cells in the circulation of rats [49].
In addition to leptin, adipose tissue produces other adipokines that affect immune cell
numbers in VAT, most notably monocyte chemo-attractant protein-1 (MCP-1) and IL-6 (Figure 2).
MCP-1 is a potent chemokine that recruits monocytes and its levels are increased in the serum of
DM2 patients [50]. Mice deficient for MCP-1 have a significant reduction in adipose tissue
inflammation and a delayed onset of insulin resistance in response to DIO [37]. The role of IL-6 in
obesity appears to be more complex and shares parallels with IL-33; IL-6 is generally assumed to be a
pro-inflammatory mediator and its levels have been shown to be significantly increased in obese
individuals [51]. The IL-6 receptor consists of the gp130 and IL-6R subunits. Although IL-6 can bind
directly to this receptor complex, IL-6 can also be bound to a soluble form of gp130 and be trans-
presented to cells that express only the IL-6R subunit [52]. Trans-presentation of IL-6 bound to
gp130 has been shown to be crucial for the accumulation of macrophages in adipose tissue [53].
Paradoxically, IL-6-deficient mice develop late-onset glucose tolerance [54]. IL-6 has been shown to
stimulate IL-4R expression on macrophages, thereby promoting M2 polarization of these cells [10],
and macrophages express high levels of the IL-6 receptor. Therefore, similarly to IL-33, under
homeostatic conditions IL-6 appears to function as an anti-inflammatory mediator by preventing M1
macrophage formation. In response to DIO, IL-6 levels increase [55], allowing other, pro-
inflammatory cells to be activated by this cytokine. Therefore, the effects of IL-6 on the development
of DM2 appear to be time- and concentration dependent.
In addition to the factors mentioned above, there is an increasingly large number of
adipokines that has been shown to mediate immune cell function in adipose tissue, such as resistin,
This article is protected by copyright. All rights reserved. 10
visfatin and apelin [56]. The exact role of these molecules in the promotion or inhibition of obesity-
induced VAT-inflammation and the development of insulin resistance is mostly unclear. However,
the sheer number of immune-modulatory molecules derived from adipose tissue clearly illustrates
the extensive communication of fat with its tissue-resident immune system.
The “Big Bang: Initiation of inflammation in VAT
Whereas the accumulation of immune cells predisposes an organ to tissue inflammation, in
itself this local accumulation is insufficient for the activation of the immune system. A key event in
the induction of obesity-induced VAT inflammation and development of insulin resistance appears to
be the polarization of macrophages from an M2 to a pro-inflammatory M1-like state [9, 13]. In
evidence of this, depletion of M1 macrophages in both humans and mice has been shown to cause
significant improvement of systemic insulin resistance [13, 57]. Pro-inflammatory licensing of
macrophages is therefore crucial for the development of VAT inflammation, as they appear to be the
primary source of pro-inflammatory cytokines that are found in the circulation of subjects with
diabetes [55]. In addition, macrophages potently recruit T cells via the production of chemokines
such as CXCL16 [58], a molecule of which the expression is greatly increased in adipose tissue in
response to obesity [59]. Thus, macrophages contribute both directly and indirectly to the excretion
of pro-inflammatory cytokines.
Systemic sources of immune activation
Various stimuli for M1 macrophage priming have been proposed, including free fatty acids
(FFAs) and microbial components [23, 60]. FFAs are normal components of our food, but they are
also the vehicle by which triacylglycerol stored in adipose tissue is transported through the
bloodstream [61]. FFAs are used as a source of fuel by many tissues, including skeletal muscle and
the brain [61]. Uncontrolled (i.e. non-treated) diabetes patients have increased amounts of FFAs in
circulation [62]. Importantly, FFAs were shown to be able to stimulate TLR4 on macrophages, but
only when bound to the protein carrier Fetuin-A [23, 63]. Palmitate, a FFA found in many food
sources, including palm oil, cheese and meat, has been shown to drive activation of the
inflammasome, the protein complex responsible for the formation of pro-inflammatory cytokines IL-
-18 [64]. Recently, the role of FFAs as initiators of inflammation is being questioned.
This article is protected by copyright. All rights reserved. 11
In large cohort studies of obese non-diabetic individuals, the levels of FFAs in the serum did not
correlate with BMI [65, 66]. Importantly, meta-analysis shows that serum FFA levels are not
predictive for the formation of DM2 [61]. FFA uptake from the circulation is controlled by insulin and
the increase of FFA levels in the blood of uncontrolled DM2 patients is therefore thought to be a
result of insulin resistance rather than its cause [61]. Indeed, well-controlled DM2 patients do not
have higher serum levels of FFA than healthy subjects [61, 67]. Notably, in vitro studies demonstrate
that FFAs can only induce pro-inflammatory cytokine excretion in macrophages when they are first
primed with LPS or IFN- [64]. Thus, before a free fatty acid can function as a pro-inflammatory
mediator, polarization of ATMs is first required. This observation in turn triggered the hypothesis
that obesity-induced gut dysbiosis drives adipose tissue inflammation, through the leakage of
microbial components (or pathogen-associated molecular patterns (PAMPs)) into the blood stream
(Figure 2). Whereas large-scale, metagenome-wide association studies on microbial gut DNA
revealed only mild dysbiosis in the intestines of DM2 patients [68], several micro-organisms could
indeed be associated with markers of metabolic syndrome [69]. Animal studies indicate that a HFD
increases endotoxin levels in the circulation [60, 70]. However, this could only be achieved when
food with a very high fat content was used, whereas more commonly used high-fat diets failed to do
so [60]. Nevertheless, studies in humans indicate that HFDs acutely increase systemic endotoxin
levels [71]. More importantly, a large prospective cohort study showed that systemic endotoxin
levels are a risk factor for the development of DM2 [72]. Thus, there is clear evidence that systemic
factors, especially diet-induced endotoxemia, play a role in the development of insulin resistance.
Whether and how these factors affect chronic systemic inflammation has yet to be elucidated.
Local sources of VAT inflammation
Systemic endotoxemia does not explain, however, why VAT macrophages specifically would
be primed towards an M1 phenotype. Rather, a local trigger is required to initiate macrophage
polarization in particularly this tissue. IFN- is a potent inducer of M1 polarization [73]. Mice
deficient for this cytokine have reduced M1 macrophage accumulation in the VAT and improved
insulin sensitivity in response to HFD feeding [74-76]. Therefore, obesity-induced IFN- production
has been suggested to be an important initiating event in VAT inflammation.
NK cells are   sentinels and browse tissues looking for signs of cellular stress,
activating the immune system in response to viral infection or oncogenic transformation. NK cells
This article is protected by copyright. All rights reserved. 12
are armed with a broad repertoire of activating and inhibitory receptors that recognize self-ligands
on target cells. In the absence of inhibi    and/or in the presence of
   cells are triggered to induce a cytolytic response, or produce
cytokines such as IFN- [77]. In several models, NK cells have been shown to drive M1 macrophage
polarization through the production of IFN- [73, 78].
Hypertrophy is a well-known stress factor for adipocytes, leading to micro-hypoxia, ER stress
and extracellular matrix confinement [79, 80]. Indeed, we have observed that in response to HFD
feeding, murine adipocytes of the visceral fat induce expression of stress ligands which bind to the
activating NK-cell receptor NKp46 (NCR1 in mice) (Figure 2) [7]. Early after initiation of HFD feeding,
the number of IFN--producing cells increase, and in particular the number of NK cells [7]. Antibody-
mediated depletion of NK cells or deficiency of NCR1 in mice was shown to result in a significant
reduction of M1-polarized ATMs upon HFD feeding, whereas the total number of ATMs was not
greatly affected [7, 81]. Furthermore, the development of diet-induced insulin resistance could be
delayed by preventing NK-cell activation using soluble NCR1 [7]. These findings are in line with
several recent studies in humans and mice. VAT from obese humans shows a significant increase in
the number of NK cells compared with that in subcutaneous fat, and these cells produce higher
levels of IFN-[76]. In addition, human adipocytes can stimulate NK cells to produce IFN- in vitro
[82].
In addition to NK cells, T cells are an important source of IFN-[7, 19]. Both CD4+ and
CD8+ T-cell numbers strongly increase in response to DIO and ultimately become the most prevalent
lymphocytes in VAT, albeit with delayed kinetics compared to NK cells [7, 83]. In the first weeks after
DIO, CD4+ T cells retain a TH2 phenotype and contribute to the inhibition of VAT-inflammation [19].
CD8+ T cells on the other hand, have been shown to recruit macrophages through the production of
MCP-1 and their deletion in mice results in a significant decrease in systemic inflammation and
insulin resistance upon DIO [84]. It is currently unclear whether CD4+ and CD8+ T cells are activated
in VAT through engagement of their T-cell receptor (TCR), or whether they are recruited in a
chemokine-dependent fashion, independent of TCR engagement. Obese, but not lean adipose tissue
was shown to be able to induce proliferation of CD8+ T cells in vitro [84]. However, whether this is
also the way via which CD8+ T-cell numbers increase in obese VAT in vivo remains to be investigated.
Finally, NKT cells can be a source of IFN- in VAT. Whereas iNKT cells initially have an anti-
inflammatory role in VAT through the production of IL-4 and IL-13, in response to HFD the number of
TNF and IFN--producing NKT cells strongly increases [7, 83]. Whether and how these cells
This article is protected by copyright. All rights reserved. 13
contribute to initiation of VAT inflammation is still a matter of debate, especially since people using
the same mouse models deficient for iNKT cells found opposing effects on the development of
insulin resistance following DIO [14, 83].
In summary, the increase in macrophage cell numbers in VAT in response to HFD feeding is
promoted by various factors, both derived from local and systemic sources. However, in order to
achieve their full pro-inflammatory potential, licensing of ATMs by NK cells through immune cell
derived cytokines such as IFN-Figure 2). The observation that IFN- deficient mice still
develop insulin resistance in response to HFD feeding, though delayed compared to wild type
animals, also indicates that this cytokine is not the only factor that drives VAT inflammation [75].
An additional layer of complexity has recently been added with the surprising observation
that recruitment of pro-inflammatory macrophages in VAT is not absolutely required for the
development of obesity-induced insulin resistance. Mice in which IL-6 trans signaling was impaired
showed reduced recruitment of macrophages into the VAT, but still developed glucose intolerance
[53]. Possibly, redundancy between pro-inflammatory mediators ensures recruitment of pro-
inflammatory cells to the VAT even in the absence of macrophages.
Inflation of inflammation in the VAT
The later stages of adipose tissue inflammation are well studied and have been elaborately
described in excellent reviews [2, 15, 85]. Therefore, we will describe this phase only briefly here.
Priming of macrophages in adipose tissue makes them susceptible to a range of pro-inflammatory
stimuli that are associated with obesity, described below. In response to obesity, adipocytes induce
expression of enzymes that convert arachidonic acid into pro-inflammatory mediators, including
leukotrienes [39, 86]. Deficiency in key components of leukotriene biology has been shown to
prevent cytokine secretion by macrophages and obesity-induced insulin resistance [39, 86]. In
addition, obesity has been shown to induce necrotic cell death of adipocytes as a result of micro-
hypoxia [79]. Necrosis results in the release of danger-associated molecular patterns (DAMPs), such
as double-stranded DNA and heat shock proteins, which drives macrophages to produce pro-
inflammatory cytokines [87]. Advanced obesity is therefore characterized by adipocytes in VAT that
are surrounded by a ring of pro-inflammatory macrophages, known as crown-like structures [88]. In
addition, priming makes murine macrophages susceptible to activation by FFAs [63]. These pro-
inflammatory stimuli drive the activation of the JNK and NF-B signaling cascades and of the
This article is protected by copyright. All rights reserved. 14
inflammasome in macrophages [87, 89, 90]. The inflammasome is a protein complex, which converts
pro-IL-1 and pro-IL-18 into IL--18, both potent activators of the immune system[91]. Either
genetic deficiency or chemical inhibition for components of the JNK, NF-B or inflammasome
pathways has been shown to strongly reduce obesity-induced insulin resistance in humans and mice
[87, 89, 90].
The activation of macrophages in VAT and the generation of an inflammatory environment
in this tissue subsequently drives the recruitment of a plethora of pro-inflammatory cells, mostly
with a TH1 signature. TH1 effector CD4 T cells, CD8 T cells and B cells most strongly increase during
the later stages of adipose tissue inflammation [2, 15, 19, 85]. Not only are these cells important
sources of IFN- that further drive ATM polarization, they also contribute to chronic systemic
inflammation by producing pro-inflammatory cytokines such as TNF (Figure 2). Ultimately, it is the
chronic leakage of these pro-inflammatory cytokines into the blood stream that forms a strong risk
factor for the development of systemic insulin resistance by dampening the signaling capacity of the
insulin receptor in local tissues [3].
Concluding remarks
Although the immunological complexity of VAT inflammation is becoming increasingly clear,
many questions remain unanswered. Subcutaneous adipose tissue is comparable in many aspects to
its visceral counterpart, including the capacity to produce leptin and adiponectin [92], yet it
accumulates much less pro-inflammatory macrophages in response to DIO [93]. Adipose tissue
stroma is an important source of adipokines and has been shown to activate CD8+ T cells in HFD-fed
animals [84, 92]. Therefore, differences between the connective tissues that support visceral and
subcutaneous adipocytes are likely to be important. In addition, the chronic systemic presence of
pro-inflammatory mediators cannot in itself explain the development of insulin resistance. Whereas
some inflammatory diseases, such as psoriasis, ulcerative colitis and systemic vasculitis are
ry
arthritis and systemic autoimmune disorders (e.g SLE, scleroderma) do not [94]. Clearly,
communication between metabolic and immunological regulators is required for the development
of DM2.
Finally, a better understanding of the underlying mechanisms behind adipose tissue
inflammation holds great promise as a target for future therapeutic intervention. Current clinical
This article is protected by copyright. All rights reserved. 15
practice mainly focuses on the reduction of blood glucose levels in DM2 patients[95], which is a
consequence of insulin resistance, whereas obesity-induced VAT inflammation is crucial for the
development this disease. Several phase II and III clinical trials have been initiated to inhibit key
immunological processes of VAT inflammation in DM2 patients, such as NF- , IL-
function or arachidonic acid metabolism, with promising results [85]. Thus, targeting of
inflammatory processes in VAT of obese patients appears to be a promising future strategy for
prophylaxis against diabetes development.
Acknowledgments:
This work is supported by the European Foundation for the Study of Diabetes (New Horizons
Program), the Unity through Knowledge Fund (15/13 to B.P.), the University of Rijeka (13.06.1.1.03
to B.P.), the European Social Fund - ES (HR.3.2.01-0263 to B.P.), the Netherlands Organization for
Scientific Research (91614029 to F.M.W.) and the European Commission (PCIG14-GA-2013-630827
to F.M.W.).
Conflict of interest
The authors declare no financial or commercial conflict of interest.
1 Stevens, G. A., Singh, G. M., Lu, Y., Danaei, G., Lin, J. K., Finucane, M. M., Bahalim, A. N.,
McIntire, R. K., Gutierrez, H. R., Cowan, M., Paciorek, C. J., Farzadfar, F., Riley, L. and Ezzati,
M., National, regional, and global trends in adult overweight and obesity prevalences. Popul
Health Metr 2012. 10: 22.
2 Johnson, A. R., Milner, J. J. and Makowski, L., The inflammation highway: metabolism
accelerates inflammatory traffic in obesity. Immunol Rev 2012. 249: 218-238.
3 Rui, L., Yuan, M., Frantz, D., Shoelson, S. and White, M. F., SOCS-1 and SOCS-3 block insulin
signaling by ubiquitin-mediated degradation of IRS1 and IRS2. J Biol Chem 2002. 277: 42394-
42398.
4 Nedergaard, J., Bengtsson, T. and Cannon, B., Unexpected evidence for active brown
adipose tissue in adult humans. Am J Physiol Endocrinol Metab 2007. 293: E444-452.
5 Ronti, T., Lupattelli, G. and Mannarino, E., The endocrine function of adipose tissue: an
update. Clin Endocrinol (Oxf) 2006. 64: 355-365.
6 Sanchez-Gurmaches, J. and Guertin, D. A., Adipocytes arise from multiple lineages that are
heterogeneously and dynamically distributed. Nat Commun 2014. 5: 4099.
7 Wensveen, F. M., Jelencic, V., Valentic, S., Sestan, M., Wensveen, T. T., Theurich, S.,
Glasner, A., Mendrila, D., Stimac, D., Wunderlich, F. T., Bruning, J. C., Mandelboim, O. and
This article is protected by copyright. All rights reserved. 16
Polic, B., NK cells link obesity-induced adipose stress to inflammation and insulin resistance.
Nat Immunol 2015. 16: 376-385.
8 Zhu, J., Yamane, H. and Paul, W. E., Differentiation of effector CD4 T cell populations (*).
Annu Rev Immunol 2010. 28: 445-489.
9 Lumeng, C. N., Bodzin, J. L. and Saltiel, A. R., Obesity induces a phenotypic switch in adipose
tissue macrophage polarization. J Clin Invest 2007. 117: 175-184.
10 Mauer, J., Chaurasia, B., Goldau, J., Vogt, M. C., Ruud, J., Nguyen, K. D., Theurich, S.,
Hausen, A. C., Schmitz, J., Bronneke, H. S., Estevez, E., Allen, T. L., Mesaros, A., Partridge,
L., Febbraio, M. A., Chawla, A., Wunderlich, F. T. and Bruning, J. C., Signaling by IL-6
promotes alternative activation of macrophages to limit endotoxemia and obesity-
associated resistance to insulin. Nat Immunol 2014. 15: 423-430.
11 Wu, D., Molofsky, A. B., Liang, H. E., Ricardo-Gonzalez, R. R., Jouihan, H. A., Bando, J. K.,
Chawla, A. and Locksley, R. M., Eosinophils sustain adipose alternatively activated
macrophages associated with glucose homeostasis. Science 2011. 332: 243-247.
12 Molofsky, A. B., Nussbaum, J. C., Liang, H. E., Van Dyken, S. J., Cheng, L. E., Mohapatra, A.,
Chawla, A. and Locksley, R. M., Innate lymphoid type 2 cells sustain visceral adipose tissue
eosinophils and alternatively activated macrophages. J Exp Med 2013. 210: 535-549.
13 Patsouris, D., Li, P. P., Thapar, D., Chapman, J., Olefsky, J. M. and Neels, J. G., Ablation of
CD11c-positive cells normalizes insulin sensitivity in obese insulin resistant animals. Cell
Metab 2008. 8: 301-309.
14 Schipper, H. S., Rakhshandehroo, M., van de Graaf, S. F., Venken, K., Koppen, A., Stienstra,
R., Prop, S., Meerding, J., Hamers, N., Besra, G., Boon, L., Nieuwenhuis, E. E., Elewaut, D.,
Prakken, B., Kersten, S., Boes, M. and Kalkhoven, E., Natural killer T cells in adipose tissue
prevent insulin resistance. J Clin Invest 2012. 122: 3343-3354.
15 Mathis, D., Immunological goings-on in visceral adipose tissue. Cell Metab 2013. 17: 851-
859.
16 Vasanthakumar, A., Moro, K., Xin, A., Liao, Y., Gloury, R., Kawamoto, S., Fagarasan, S.,
Mielke, L. A., Afshar-Sterle, S., Masters, S. L., Nakae, S., Saito, H., Wentworth, J. M., Li, P.,
Liao, W., Leonard, W. J., Smyth, G. K., Shi, W., Nutt, S. L., Koyasu, S. and Kallies, A., The
transcriptional regulators IRF4, BATF and IL-33 orchestrate development and maintenance of
adipose tissue-resident regulatory T cells. Nat Immunol 2015. 16: 276-285.
17 Cipolletta, D., Feuerer, M., Li, A., Kamei, N., Lee, J., Shoelson, S. E., Benoist, C. and Mathis,
D., PPAR-gamma is a major driver of the accumulation and phenotype of adipose tissue Treg
cells. Nature 2012. 486: 549-553.
18 Feuerer, M., Herrero, L., Cipolletta, D., Naaz, A., Wong, J., Nayer, A., Lee, J., Goldfine, A. B.,
Benoist, C., Shoelson, S. and Mathis, D., Lean, but not obese, fat is enriched for a unique
population of regulatory T cells that affect metabolic parameters. Nat Med 2009. 15: 930-
939.
19 Winer, S., Chan, Y., Paltser, G., Truong, D., Tsui, H., Bahrami, J., Dorfman, R., Wang, Y.,
Zielenski, J., Mastronardi, F., Maezawa, Y., Drucker, D. J., Engleman, E., Winer, D. and
Dosch, H. M., Normalization of obesity-associated insulin resistance through
immunotherapy. Nat Med 2009. 15: 921-929.
20 Ouchi, N., Parker, J. L., Lugus, J. J. and Walsh, K., Adipokines in inflammation and metabolic
disease. Nat Rev Immunol 2011. 11: 85-97.
21 Yamauchi, T., Kamon, J., Waki, H., Terauchi, Y., Kubota, N., Hara, K., Mori, Y., Ide, T.,
Murakami, K., Tsuboyama-Kasaoka, N., Ezaki, O., Akanuma, Y., Gavrilova, O., Vinson, C.,
Reitman, M. L., Kagechika, H., Shudo, K., Yoda, M., Nakano, Y., Tobe, K., Nagai, R., Kimura,
S., Tomita, M., Froguel, P. and Kadowaki, T., The fat-derived hormone adiponectin reverses
insulin resistance associated with both lipoatrophy and obesity. Nat Med 2001. 7: 941-946.
This article is protected by copyright. All rights reserved. 17
22 Barnes, M. A., Carson, M. J. and Nair, M. G., Non-traditional cytokines: How catecholamines
and adipokines influence macrophages in immunity, metabolism and the central nervous
system. Cytokine 2015. 72: 210-219.
23 Shi, H., Kokoeva, M. V., Inouye, K., Tzameli, I., Yin, H. and Flier, J. S., TLR4 links innate
immunity and fatty acid-induced insulin resistance. J Clin Invest 2006. 116: 3015-3025.
24 Liu, J., Divoux, A., Sun, J., Zhang, J., Clement, K., Glickman, J. N., Sukhova, G. K., Wolters, P.
J., Du, J., Gorgun, C. Z., Doria, A., Libby, P., Blumberg, R. S., Kahn, B. B., Hotamisligil, G. S.
and Shi, G. P., Genetic deficiency and pharmacological stabilization of mast cells reduce diet-
induced obesity and diabetes in mice. Nat Med 2009. 15: 940-945.
25 Zuniga, L. A., Shen, W. J., Joyce-Shaikh, B., Pyatnova, E. A., Richards, A. G., Thom, C.,
Andrade, S. M., Cua, D. J., Kraemer, F. B. and Butcher, E. C., IL-17 regulates adipogenesis,
glucose homeostasis, and obesity. J Immunol 2010. 185: 6947-6959.
26 Lynch, L., Nowak, M., Varghese, B., Clark, J., Hogan, A. E., Toxavidis, V., Balk, S. P., O'Shea,
D., O'Farrelly, C. and Exley, M. A., Adipose tissue invariant NKT cells protect against diet-
induced obesity and metabolic disorder through regulatory cytokine production. Immunity
2012. 37: 574-587.
27 Ohmura, K., Ishimori, N., Ohmura, Y., Tokuhara, S., Nozawa, A., Horii, S., Andoh, Y., Fujii,
S., Iwabuchi, K., Onoe, K. and Tsutsui, H., Natural killer T cells are involved in adipose tissues
inflammation and glucose intolerance in diet-induced obese mice. Arterioscler Thromb Vasc
Biol 2010. 30: 193-199.
28 Miller, A. M., Asquith, D. L., Hueber, A. J., Anderson, L. A., Holmes, W. M., McKenzie, A. N.,
Xu, D., Sattar, N., McInnes, I. B. and Liew, F. Y., Interleukin-33 induces protective effects in
adipose tissue inflammation during obesity in mice. Circ Res 2010. 107: 650-658.
29 Molofsky, A. B., Savage, A. K. and Locksley, R. M., Interleukin-33 in Tissue Homeostasis,
Injury, and Inflammation. Immunity 2015. 42: 1005-1019.
30 Pichery, M., Mirey, E., Mercier, P., Lefrancais, E., Dujardin, A., Ortega, N. and Girard, J. P.,
Endogenous IL-33 is highly expressed in mouse epithelial barrier tissues, lymphoid organs,
brain, embryos, and inflamed tissues: in situ analysis using a novel Il-33-LacZ gene trap
reporter strain. J Immunol 2012. 188: 3488-3495.
31 Hashiguchi, M., Kashiwakura, Y., Kojima, H., Kobayashi, A., Kanno, Y. and Kobata, T., IL-33
activates eosinophils of visceral adipose tissue both directly and via innate lymphoid cells.
Eur J Immunol 2015. 45: 876-885.
32 Brestoff, J. R., Kim, B. S., Saenz, S. A., Stine, R. R., Monticelli, L. A., Sonnenberg, G. F.,
Thome, J. J., Farber, D. L., Lutfy, K., Seale, P. and Artis, D., Group 2 innate lymphoid cells
promote beiging of white adipose tissue and limit obesity. Nature 2015. 519: 242-246.
33 Lee, M. W., Odegaard, J. I., Mukundan, L., Qiu, Y., Molofsky, A. B., Nussbaum, J. C., Yun, K.,
Locksley, R. M. and Chawla, A., Activated type 2 innate lymphoid cells regulate beige fat
biogenesis. Cell 2015. 160: 74-87.
34 Talukdar, S., Oh da, Y., Bandyopadhyay, G., Li, D., Xu, J., McNelis, J., Lu, M., Li, P., Yan, Q.,
Zhu, Y., Ofrecio, J., Lin, M., Brenner, M. B. and Olefsky, J. M., Neutrophils mediate insulin
resistance in mice fed a high-fat diet through secreted elastase. Nat Med 2012. 18: 1407-
1412.
35 Tam, C. S., Viardot, A., Clement, K., Tordjman, J., Tonks, K., Greenfield, J. R., Campbell, L.
V., Samocha-Bonet, D. and Heilbronn, L. K., Short-term overfeeding may induce peripheral
insulin resistance without altering subcutaneous adipose tissue macrophages in humans.
Diabetes 2010. 59: 2164-2170.
36 Jeffery, E., Church, C. D., Holtrup, B., Colman, L. and Rodeheffer, M. S., Rapid depot-specific
activation of adipocyte precursor cells at the onset of obesity. Nat Cell Biol 2015.
37 Kanda, H., Tateya, S., Tamori, Y., Kotani, K., Hiasa, K., Kitazawa, R., Kitazawa, S., Miyachi,
H., Maeda, S., Egashira, K. and Kasuga, M., MCP-1 contributes to macrophage infiltration
This article is protected by copyright. All rights reserved. 18
into adipose tissue, insulin resistance, and hepatic steatosis in obesity. J Clin Invest 2006.
116: 1494-1505.
38 Amano, S. U., Cohen, J. L., Vangala, P., Tencerova, M., Nicoloro, S. M., Yawe, J. C., Shen, Y.,
Czech, M. P. and Aouadi, M., Local proliferation of macrophages contributes to obesity-
associated adipose tissue inflammation. Cell Metab 2014. 19: 162-171.
39 Li, P., Oh da, Y., Bandyopadhyay, G., Lagakos, W. S., Talukdar, S., Osborn, O., Johnson, A.,
Chung, H., Mayoral, R., Maris, M., Ofrecio, J. M., Taguchi, S., Lu, M. and Olefsky, J. M., LTB4
promotes insulin resistance in obese mice by acting on macrophages, hepatocytes and
myocytes. Nat Med 2015. 21: 239-247.
40 Molofsky, A. B., Van Gool, F., Liang, H. E., Van Dyken, S. J., Nussbaum, J. C., Lee, J.,
Bluestone, J. A. and Locksley, R. M., Interleukin-33 and Interferon-gamma Counter-Regulate
Group 2 Innate Lymphoid Cell Activation during Immune Perturbation. Immunity 2015.
41 Wang, Q. A., Tao, C., Gupta, R. K. and Scherer, P. E., Tracking adipogenesis during white
adipose tissue development, expansion and regeneration. Nat Med 2013. 19: 1338-1344.
42 Zhang, Y., Proenca, R., Maffei, M., Barone, M., Leopold, L. and Friedman, J. M., Positional
cloning of the mouse obese gene and its human homologue. Nature 1994. 372: 425-432.
43 Banks, A. S., Davis, S. M., Bates, S. H. and Myers, M. G., Jr., Activation of downstream
signals by the long form of the leptin receptor. J Biol Chem 2000. 275: 14563-14572.
44 Moraes-Vieira, P. M., Larocca, R. A., Bassi, E. J., Peron, J. P., Andrade-Oliveira, V., Wasinski,
F., Araujo, R., Thornley, T., Quintana, F. J., Basso, A. S., Strom, T. B. and Camara, N. O.,
Leptin deficiency impairs maturation of dendritic cells and enhances induction of regulatory
T and Th17 cells. Eur J Immunol 2014. 44: 794-806.
45 La Cava, A. and Matarese, G., The weight of leptin in immunity. Nat Rev Immunol 2004. 4:
371-379.
46 Papathanassoglou, E., El-Haschimi, K., Li, X. C., Matarese, G., Strom, T. and Mantzoros, C.,
Leptin receptor expression and signaling in lymphocytes: kinetics during lymphocyte
activation, role in lymphocyte survival, and response to high fat diet in mice. J Immunol
2006. 176: 7745-7752.
47 Tian, Z., Sun, R., Wei, H. and Gao, B., Impaired natural killer (NK) cell activity in leptin
receptor deficient mice: leptin as a critical regulator in NK cell development and activation.
Biochem Biophys Res Commun 2002. 298: 297-302.
48 Lo, C. K., Lam, Q. L., Yang, M., Ko, K. H., Sun, L., Ma, R., Wang, S., Xu, H., Tam, S., Wu, C. Y.,
Zheng, B. J. and Lu, L., Leptin signaling protects NK cells from apoptosis during development
in mouse bone marrow. Cell Mol Immunol 2009. 6: 353-360.
49 Haas, P., Straub, R. H., Bedoui, S. and Nave, H., Peripheral but not central leptin treatment
increases numbers of circulating NK cells, granulocytes and specific monocyte
subpopulations in non-endotoxaemic lean and obese LEW-rats. Regul Pept 2008. 151: 26-34.
50 Zietz, B., Buchler, C., Herfarth, H., Muller-Ladner, U., Spiegel, D., Scholmerich, J. and
Schaffler, A., Caucasian patients with type 2 diabetes mellitus have elevated levels of
monocyte chemoattractant protein-1 that are not influenced by the -2518 A-->G promoter
polymorphism. Diabetes Obes Metab 2005. 7: 570-578.
51 Eder, K., Baffy, N., Falus, A. and Fulop, A. K., The major inflammatory mediator interleukin-6
and obesity. Inflamm Res 2009. 58: 727-736.
52 Scheller, J., Chalaris, A., Schmidt-Arras, D. and Rose-John, S., The pro- and anti-
inflammatory properties of the cytokine interleukin-6. Biochim Biophys Acta 2011. 1813:
878-888.
53 Kraakman, M. J., Kammoun, H. L., Allen, T. L., Deswaerte, V., Henstridge, D. C., Estevez, E.,
Matthews, V. B., Neill, B., White, D. A., Murphy, A. J., Peijs, L., Yang, C., Risis, S., Bruce, C.
R., Du, X. J., Bobik, A., Lee-Young, R. S., Kingwell, B. A., Vasanthakumar, A., Shi, W., Kallies,
A., Lancaster, G. I., Rose-John, S. and Febbraio, M. A., Blocking IL-6 trans-Signaling Prevents
This article is protected by copyright. All rights reserved. 19
High-Fat Diet-Induced Adipose Tissue Macrophage Recruitment but Does Not Improve
Insulin Resistance. Cell Metab 2015. 21: 403-416.
54 Wallenius, V., Wallenius, K., Ahren, B., Rudling, M., Carlsten, H., Dickson, S. L., Ohlsson, C.
and Jansson, J. O., Interleukin-6-deficient mice develop mature-onset obesity. Nat Med
2002. 8: 75-79.
55 Starr, M. E., Evers, B. M. and Saito, H., Age-associated increase in cytokine production
during systemic inflammation: adipose tissue as a major source of IL-6. J Gerontol A Biol Sci
Med Sci 2009. 64: 723-730.
56 Balistreri, C. R., Caruso, C. and Candore, G., The role of adipose tissue and adipokines in
obesity-related inflammatory diseases. Mediators Inflamm 2010. 2010: 802078.
57 Koppaka, S., Kehlenbrink, S., Carey, M., Li, W., Sanchez, E., Lee, D. E., Lee, H., Chen, J.,
Carrasco, E., Kishore, P., Zhang, K. and Hawkins, M., Reduced adipose tissue macrophage
content is associated with improved insulin sensitivity in thiazolidinedione-treated diabetic
humans. Diabetes 2013. 62: 1843-1854.
58 van der Voort, R., van Lieshout, A. W., Toonen, L. W., Sloetjes, A. W., van den Berg, W. B.,
Figdor, C. G., Radstake, T. R. and Adema, G. J., Elevated CXCL16 expression by synovial
macrophages recruits memory T cells into rheumatoid joints. Arthritis Rheum 2005. 52:
1381-1391.
59 Kurki, E., Shi, J., Martonen, E., Finckenberg, P. and Mervaala, E., Distinct effects of calorie
restriction on adipose tissue cytokine and angiogenesis profiles in obese and lean mice. Nutr
Metab (Lond) 2012. 9: 64.
60 Cani, P. D., Amar, J., Iglesias, M. A., Poggi, M., Knauf, C., Bastelica, D., Neyrinck, A. M.,
Fava, F., Tuohy, K. M., Chabo, C., Waget, A., Delmee, E., Cousin, B., Sulpice, T., Chamontin,
B., Ferrieres, J., Tanti, J. F., Gibson, G. R., Casteilla, L., Delzenne, N. M., Alessi, M. C. and
Burcelin, R., Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 2007.
56: 1761-1772.
61 Karpe, F., Dickmann, J. R. and Frayn, K. N., Fatty acids, obesity, and insulin resistance: time
for a reevaluation. Diabetes 2011. 60: 2441-2449.
62 Reaven, G. M., Hollenbeck, C., Jeng, C. Y., Wu, M. S. and Chen, Y. D., Measurement of
plasma glucose, free fatty acid, lactate, and insulin for 24 h in patients with NIDDM. Diabetes
1988. 37: 1020-1024.
63 Pal, D., Dasgupta, S., Kundu, R., Maitra, S., Das, G., Mukhopadhyay, S., Ray, S., Majumdar,
S. S. and Bhattacharya, S., Fetuin-A acts as an endogenous ligand of TLR4 to promote lipid-
induced insulin resistance. Nat Med 2012. 18: 1279-1285.
64 Wen, H., Gris, D., Lei, Y., Jha, S., Zhang, L., Huang, M. T., Brickey, W. J. and Ting, J. P., Fatty
acid-induced NLRP3-ASC inflammasome activation interferes with insulin signaling. Nat
Immunol 2011. 12: 408-415.
65 Charles, M. A., Fontbonne, A., Thibult, N., Claude, J. R., Warnet, J. M., Rosselin, G.,
Ducimetiere, P. and Eschwege, E., High plasma nonesterified fatty acids are predictive of
cancer mortality but not of coronary heart disease mortality: results from the Paris
Prospective Study. Am J Epidemiol 2001. 153: 292-298.
66 Byrne, C. D., Maison, P., Halsall, D., Martensz, N., Hales, C. N. and Wareham, N. J., Cross-
sectional but not longitudinal associations between non-esterified fatty acid levels and
glucose intolerance and other features of the metabolic syndrome. Diabet Med 1999. 16:
1007-1015.
67 Adams, J. M., 2nd, Pratipanawatr, T., Berria, R., Wang, E., DeFronzo, R. A., Sullards, M. C.
and Mandarino, L. J., Ceramide content is increased in skeletal muscle from obese insulin-
resistant humans. Diabetes 2004. 53: 25-31.
68 Qin, J., Li, Y., Cai, Z., Li, S., Zhu, J., Zhang, F., Liang, S., Zhang, W., Guan, Y., Shen, D., Peng,
Y., Zhang, D., Jie, Z., Wu, W., Qin, Y., Xue, W., Li, J., Han, L., Lu, D., Wu, P., Dai, Y., Sun, X.,
This article is protected by copyright. All rights reserved. 20
Li, Z., Tang, A., Zhong, S., Li, X., Chen, W., Xu, R., Wang, M., Feng, Q., Gong, M., Yu, J.,
Zhang, Y., Zhang, M., Hansen, T., Sanchez, G., Raes, J., Falony, G., Okuda, S., Almeida, M.,
LeChatelier, E., Renault, P., Pons, N., Batto, J. M., Zhang, Z., Chen, H., Yang, R., Zheng, W.,
Li, S., Yang, H., Wang, J., Ehrlich, S. D., Nielsen, R., Pedersen, O., Kristiansen, K. and Wang,
J., A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 2012.
490: 55-60.
69 Karlsson, F. H., Tremaroli, V., Nookaew, I., Bergstrom, G., Behre, C. J., Fagerberg, B.,
Nielsen, J. and Backhed, F., Gut metagenome in European women with normal, impaired
and diabetic glucose control. Nature 2013. 498: 99-103.
70 Cani, P. D., Bibiloni, R., Knauf, C., Waget, A., Neyrinck, A. M., Delzenne, N. M. and Burcelin,
R., Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-
fat diet-induced obesity and diabetes in mice. Diabetes 2008. 57: 1470-1481.
71 Ghanim, H., Abuaysheh, S., Sia, C. L., Korzeniewski, K., Chaudhuri, A., Fernandez-Real, J. M.
and Dandona, P., Increase in plasma endotoxin concentrations and the expression of Toll-
like receptors and suppressor of cytokine signaling-3 in mononuclear cells after a high-fat,
high-carbohydrate meal: implications for insulin resistance. Diabetes Care 2009. 32: 2281-
2287.
72 Pussinen, P. J., Havulinna, A. S., Lehto, M., Sundvall, J. and Salomaa, V., Endotoxemia is
associated with an increased risk of incident diabetes. Diabetes Care 2011. 34: 392-397.
73 Goldszmid, R. S., Caspar, P., Rivollier, A., White, S., Dzutsev, A., Hieny, S., Kelsall, B.,
Trinchieri, G. and Sher, A., NK cell-derived interferon-gamma orchestrates cellular dynamics
and the differentiation of monocytes into dendritic cells at the site of infection. Immunity
2012. 36: 1047-1059.
74 Wong, N., Fam, B. C., Cempako, G. R., Steinberg, G. R., Walder, K., Kay, T. W., Proietto, J.
and Andrikopoulos, S., Deficiency in interferon-gamma results in reduced body weight and
better glucose tolerance in mice. Endocrinology 2011. 152: 3690-3699.
75 Rocha, V. Z., Folco, E. J., Sukhova, G., Shimizu, K., Gotsman, I., Vernon, A. H. and Libby, P.,
Interferon-gamma, a Th1 cytokine, regulates fat inflammation: a role for adaptive immunity
in obesity. Circ Res 2008. 103: 467-476.
76 O'Rourke, R. W., White, A. E., Metcalf, M. D., Winters, B. R., Diggs, B. S., Zhu, X. and Marks,
D. L., Systemic inflammation and insulin sensitivity in obese IFN-gamma knockout mice.
Metabolism 2012. 61: 1152-1161.
77 Lanier, L. L., Up on the tightrope: natural killer cell activation and inhibition. Nat Immunol
2008. 9: 495-502.
78 Dungan, L. S., McGuinness, N. C., Boon, L., Lynch, M. A. and Mills, K. H., Innate IFN-gamma
promotes development of experimental autoimmune encephalomyelitis: a role for NK cells
and M1 macrophages. Eur J Immunol 2014. 44: 2903-2917.
79 Ye, J., Gao, Z., Yin, J. and He, Q., Hypoxia is a potential risk factor for chronic inflammation
and adiponectin reduction in adipose tissue of ob/ob and dietary obese mice. Am J Physiol
Endocrinol Metab 2007. 293: E1118-1128.
80 Khan, T., Muise, E. S., Iyengar, P., Wang, Z. V., Chandalia, M., Abate, N., Zhang, B. B.,
Bonaldo, P., Chua, S. and Scherer, P. E., Metabolic dysregulation and adipose tissue fibrosis:
role of collagen VI. Mol Cell Biol 2009. 29: 1575-1591.
81 O'Rourke, R. W., Meyer, K. A., Neeley, C. K., Gaston, G. D., Sekhri, P., Szumowski, M.,
Zamarron, B., Lumeng, C. N. and Marks, D. L., Systemic NK cell ablation attenuates intra-
abdominal adipose tissue macrophage infiltration in murine obesity. Obesity (Silver Spring)
2014. 22: 2109-2114.
82 Huebner, L., Engeli, S., Wrann, C. D., Goudeva, L., Laue, T. and Kielstein, H., Human NK cell
subset functions are differentially affected by adipokines. PLoS One 2013. 8: e75703.
This article is protected by copyright. All rights reserved. 21
83 Wu, L., Parekh, V. V., Gabriel, C. L., Bracy, D. P., Marks-Shulman, P. A., Tamboli, R. A., Kim,
S., Mendez-Fernandez, Y. V., Besra, G. S., Lomenick, J. P., Williams, B., Wasserman, D. H.
and Van Kaer, L., Activation of invariant natural killer T cells by lipid excess promotes tissue
inflammation, insulin resistance, and hepatic steatosis in obese mice. Proc Natl Acad Sci U S
A 2012. 109: E1143-1152.
84 Nishimura, S., Manabe, I., Nagasaki, M., Eto, K., Yamashita, H., Ohsugi, M., Otsu, M., Hara,
K., Ueki, K., Sugiura, S., Yoshimura, K., Kadowaki, T. and Nagai, R., CD8+ effector T cells
contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nat Med
2009. 15: 914-920.
85 Donath, M. Y. and Shoelson, S. E., Type 2 diabetes as an inflammatory disease. Nat Rev
Immunol 2011. 11: 98-107.
86 Mothe-Satney, I., Filloux, C., Amghar, H., Pons, C., Bourlier, V., Galitzky, J., Grimaldi, P. A.,
Feral, C. C., Bouloumie, A., Van Obberghen, E. and Neels, J. G., Adipocytes secrete
leukotrienes: contribution to obesity-associated inflammation and insulin resistance in mice.
Diabetes 2012. 61: 2311-2319.
87 Vandanmagsar, B., Youm, Y. H., Ravussin, A., Galgani, J. E., Stadler, K., Mynatt, R. L.,
Ravussin, E., Stephens, J. M. and Dixit, V. D., The NLRP3 inflammasome instigates obesity-
induced inflammation and insulin resistance. Nat Med 2011. 17: 179-188.
88 Weisberg, S. P., McCann, D., Desai, M., Rosenbaum, M., Leibel, R. L. and Ferrante, A. W.,
Jr., Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest 2003.
112: 1796-1808.
89 Arkan, M. C., Hevener, A. L., Greten, F. R., Maeda, S., Li, Z. W., Long, J. M., Wynshaw-Boris,
A., Poli, G., Olefsky, J. and Karin, M., IKK-beta links inflammation to obesity-induced insulin
resistance. Nat Med 2005. 11: 191-198.
90 Han, M. S., Jung, D. Y., Morel, C., Lakhani, S. A., Kim, J. K., Flavell, R. A. and Davis, R. J., JNK
expression by macrophages promotes obesity-induced insulin resistance and inflammation.
Science 2013. 339: 218-222.
91 Latz, E., Xiao, T. S. and Stutz, A., Activation and regulation of the inflammasomes. Nat Rev
Immunol 2013. 13: 397-411.
92 Fain, J. N., Madan, A. K., Hiler, M. L., Cheema, P. and Bahouth, S. W., Comparison of the
release of adipokines by adipose tissue, adipose tissue matrix, and adipocytes from visceral
and subcutaneous abdominal adipose tissues of obese humans. Endocrinology 2004. 145:
2273-2282.
93 Altintas, M. M., Azad, A., Nayer, B., Contreras, G., Zaias, J., Faul, C., Reiser, J. and Nayer, A.,
Mast cells, macrophages, and crown-like structures distinguish subcutaneous from visceral
fat in mice. J Lipid Res 2011. 52: 480-488.
94 Dregan, A., Charlton, J., Chowienczyk, P. and Gulliford, M. C., Chronic inflammatory
disorders and risk of type 2 diabetes mellitus, coronary heart disease, and stroke: a
population-based cohort study. Circulation 2014. 130: 837-844.
95 Inzucchi, S. E., Bergenstal, R. M., Buse, J. B., Diamant, M., Ferrannini, E., Nauck, M., Peters,
A. L., Tsapas, A., Wender, R. and Matthews, D. R., Management of hyperglycaemia in type 2
diabetes: a patient-centered approach. Position statement of the American Diabetes
Association (ADA) and the European Association for the Study of Diabetes (EASD).
Diabetologia 2012. 55: 1577-1596.
This article is protected by copyright. All rights reserved. 22
Figure 1. Control of adipose tissue homeostasis under lean conditions. Healthy adipose tissue is
populated by a Type 2-polarized immune system, which maintains ATMs in an M2-like state.
Dominant immune cell subsets under these conditions are eosinophils, iNKT cells and Treg cells,
which produce IL-4, IL-13 and IL-10 respectively. Adipocytes also contribute to the Type 2 immune
response through the production of adiponectin. ILC2s regulate eosinophil numbers through the
production of IL-5. Type 2 immune cells are supported by a stromal structure which promotes
immune cell viability through the production of several cytokines, most importantly IL-33. In
addition to sustaining a Type 2 immune cell environment, adipose tissue cells engage in extensive
cross-talk to (re)model adipose tissue structure and phenotype. One example is the formation of
beiging through the production of IL-4, IL-13
and Met-Enkephalin by the ILC2/eosinophil axis. NKT = Invariant-Chain Natural Killer T cell, TReg =
regulatory T cell, Eo = Eosinophil, M2 = M2 Macrophage, ILC2 = Innate Lymphocyte 2, AP = adipocyte
       Stroma = adipose tissue stroma. MetEnk = Met-
enkephalin.
M2
NKT
Treg WA
AP
Brite
IL-10
IL-33
IL-33
IL-33
IL-25
TSLP
IL-4
IL-4
IL-5
IL-13
Adiponectin
MetEnk
IL-13
Stroma
?
ILC2
Eo
This article is protected by copyright. All rights reserved. 23
Figure 2. Model for development of obesity-induced adipose tissue inflammation. In response to
HFD, adipocytes initially become hypertrophic and later hyperplastic (indicated by increase in size
and number of adipocytes respectively). This is associated with a shift in adipokine production from
adiponectin to leptin/MCP-1 and an increase in the number of ATMs, neutrophils and NK cells in
visceral fat. As obesity persists, adipocyte stress drives CD8+ T-cell and NK-cell activation through
NKp46, resulting in local production of IFN-coming from the periphery, this
locally-produced IFN-  -inflammatory M1 state. This makes these cells
sensitive to a range of pro-inflammatory stimuli, such as leukotrienes, FFA-Fetuin-A complexes and
DAMPs from necrotic adipocytes. As a result, ATMs produce pro-inflammatory cytokines, such as
TNF and IL- and recruit more pro-inflammatory cells, including CD4+ T cells, CD8+ T cells and B
cells, into the VAT to amplify the immune response. The chronic systemic presence of pro-
inflammatory cytokines derived from this response ultimately contributes to the development of
insulin resistance. M1 = M1 Macrophage, M2 = M2 Macrophage, 
cell, IFN-
DAMP = danger associated molecular pattern, TNF = tumor necrosis factor.
CIRCULATION
M2 M2
M2
M2
NK
NK
NK
M1
M1
M1
M1
NK
CD4
CD8
B
Insulin
Resistance
DAMP
TNF
IL-1β
Leukotrienes
Adiponectin
Leptin, MCP-1
Adiponectin
Leptin, MCP-1
IFNγ
CD8
PAMP
Nφ
FFA-
FetuinA
?
... Obesity has long been considered a public health problem around the world and a major factor responsible for reducing quality of life as well as increasing morbidity and mortality (Williams et al., 2015). Several studies have demonstrated that obesity is frequently accompanied by chronic low-grade inflammation marked by increased circulating levels of leptin, interleukin-6 (IL-6), tumor necrosis factor -α (TNF-α), C-reactive protein (CRP) and monocyte chemotactic protein − 1 (MCP1) (Ghigliotti et al., 2014;Wensveen et al., 2015;Williams et al., 2015), together with dysregulation in inflammation-stress feedback mechanisms (Martín-Cordero et al., 2011). Thus, obesity has come to be associated with the development of metabolic syndrome and several diseases such as dyslipidemia, type 2 diabetes mellitus (T2D), cardiovascular diseases, chronic obstructive pulmonary disease, and among others, various types of cancer, and depression (Wensveen et al., 2015;Williams et al., 2015). ...
... Several studies have demonstrated that obesity is frequently accompanied by chronic low-grade inflammation marked by increased circulating levels of leptin, interleukin-6 (IL-6), tumor necrosis factor -α (TNF-α), C-reactive protein (CRP) and monocyte chemotactic protein − 1 (MCP1) (Ghigliotti et al., 2014;Wensveen et al., 2015;Williams et al., 2015), together with dysregulation in inflammation-stress feedback mechanisms (Martín-Cordero et al., 2011). Thus, obesity has come to be associated with the development of metabolic syndrome and several diseases such as dyslipidemia, type 2 diabetes mellitus (T2D), cardiovascular diseases, chronic obstructive pulmonary disease, and among others, various types of cancer, and depression (Wensveen et al., 2015;Williams et al., 2015). ...
... Furthermore, we observed a decrease in IL-4 gene expression in the PBMCs of obese T2D individuals after the training period. It is known that IL-4 is involved in type II inflammatory response and is the key regulator of activation, growth and differentiation of lymphocyte functions (Juntilla, 2018) and plays a protective role in inflammation by suppressing proinflammatory cytokine production in, for example, TNF-α and IL-1 (Wensveen et al., 2015). Previous studies have demonstrated that IL-4 expression is increased in PBMCs of obese individuals with T2D (Brunelli et al., 2022;Dandona et al., 2014). ...
... Similarly, in humans, individuals with overweight or obesity have reduced Th2 cells, and Th2 cells in the visceral adiposity and peripheral blood are negatively associated with systemic IR and plasma highsensitivity C-reactive protein 69 . Mechanistically, Th2 cells enhance glucose utilization by producing IL-4 polarized macrophages, increase peroxisome proliferator-activated receptor gamma (PPARγ) driven GLUT4 expression 71 , and activate the mechanistic target of the rapamycin complex (mTORC)2 pathway 72 . ...
Preprint
Type 2 diabetes mellitus (T2DM) is a chronic metabolic disease characterized by hyperglycemia. T2DM is caused by various etiologies. The functional expansion of pancreatic β-cells is unable to compensate for the degree of insulin resistance (IR), resulting in a relative insulin deficiency. The onset and progression of T2DM are influenced by multiple variables, including genetics, lipid excess, oxidative stress, and inflammation. A growing body of research suggests that the components of the immune system are altered in T2DM. This suggests that T cell-mediated adaptive immunity stimulates inflammation and IR through the redistribution of cytokines, chemokines, and different T cell subsets. Metabolic inflammation is a central aspect of obesity, T2DM, and comorbidities. This review focuses on adaptive immune T cells, particularly CD4+ T cells, and examines the roles and effects of different helper T (Th) 1, Th2, Th17, Th22, and regulatory T cells (Tregs) in T2DM. Evidence for T cell activation and exhaustion in T2DM remains controversial and requires further investigation.
... However, FT-Raman spectra showed several characteristic quantitative and qualitative changes in the functional group building, especially protein structure, as seen in Fig. 1. In obese people, it was observed that during obesity, changes in the phenotypic of adipose tissue and the development of chronic low-grade inflammation [44]. This process was characterized, e.g., by increased levels of soluble pro-inflammatory factors (mainly proteins) and the activation and infiltration of immune cells into sites of inflammation [45]. ...
Article
This study utilized FT-Raman spectroscopy, linear regression models, principal component analysis (PCA), and correlation to explore the link between BMI and serum chemical structure across varying obesity stages. Biochemical, hematological, and immune-inflammatory markers were analyzed in 80 patients across three groups (overweight, obese, and morbidly obese) and healthy subjects (n = 45). Only BMI significantly varied among groups, with morbidly obese patients showing higher Cystatin c levels. In the morbidly obese patient group, a correlation was observed between BMI and 5 peaks (1270 cm−1, 1344 cm−1, 1459 cm−1, 1671 cm−1, and 2922 cm−1). FT-Raman data indicated lower amide, amino acid, and lipid levels in overweight individuals. FT-Raman data were found to correlate with BMI by more than 50 %, especially in patients with the highest degree of obesity, notably 1606 cm−1 (a 53 % fit) and 2933 cm−1 (67 % fit) suggesting strong correlations between FT-Raman data and BMI, particularly in severe obesity.
... These are the first data to report preferential NK cell migration towards the chemotactic cues of OAC patient omentum over tumour and this is in line with our previous reports of active NK cell migration to OAC patient-derived ACM, further highlighting the threat that the omentum and higher visceral adiposity pose for anti-tumour immunity in OAC 16,63 . We have previously reported compelling evidence that therapeutically precluding NK cell migration to the omentum is warranted in OAC as it would limit omental-induced reductions in NK cell viability and migratory capacity towards tumour, and minimise alterations to NK phenotype and inflammatory profile that perpetuate adipose tissue inflammation 16,[63][64][65] . We have previously confirmed that the chemokine fractalkine is a key driver of erroneous NK cell migration to OAC patient omentum 16 . ...
Article
Full-text available
Oesophagogastric adenocarcinomas (OAC) are poor prognosis, obesity-associated cancers which may benefit from natural killer (NK) cell-based immunotherapies. Cellular immunotherapies encounter two key challenges to their success in OAC, namely recruitment to extratumoural tissues such as the omentum at the expense of the tumour and an immunosuppressive tumour microenvironment (TME) which can hamper NK cell function. Herein, we examined approaches to overcome the detrimental impact of obesity on NK cells and NK cell-based immunotherapies. We have demonstrated that NK cells migrate preferentially to the chemotactic signals of OAC patient-derived omentum over tumour in an ex vivo model of immune cell migration. We have identified CX3CR1 modulation and/or tumour chemokine profile remodelling as approaches to skew NK cell migration towards tumour. We also report targetable immunosuppressive facets of the obese OAC TME which dampen NK cell function, in particular cytotoxic capabilities. These data provide insights into approaches to therapeutically overcome key challenges presented by obesity and will inform superior design of NK cell-based immunotherapies for OAC.
... Th2-type cytokines are involved in the pathogenesis of airway inflammatory diseases such as asthma. The most prominent are IL-4, IL-5, IL-6, IL-10, and IL-33 [17,41,61]. IL-4, produced by ILC2 cells and eosinophils [35], induces the expression of two anti-inflammatory cytokines by M2, TGF-β, and IL-10 [62]. ...
Article
Full-text available
Globally, obesity and asthma pose significant health challenges, with obesity being a key factor influencing asthma. Despite this, effective treatments for obese asthma, a distinct phenotype, remain elusive. Since the discovery of transient receptor potential (TRP) channels in 1969, their value as therapeutic targets for various diseases has been acknowledged. TRP channels, present in adipose tissue cells, influence fat cell heat production and the secretion of adipokines and cytokines, which are closely associated with asthma and obesity. This paper aims to investigate the mechanisms by which obesity exacerbates asthma-related inflammation and suggests that targeting TRP channels in adipose tissue could potentially suppress obese asthma and offer novel insights into its treatment.
Preprint
Obesity has been associated with dysbiosis, but innate mechanisms linking intestinal epithelial cell subsets and obesity remain poorly understood. Using mice lacking Paneth cells (Sox9 ΔIEC mice), small intestinal epithelial cells specialized in the production of antimicrobial products and cytokines, we show that dysbiosis alone does not induce obesity or metabolic disorders. Loss of Paneth cells reduced ILC3 and increased ILC2 numbers in the intestinal lamina propria. High-fat diet (HFD) induced higher weight gain and more severe metabolic disorders in Sox9 ΔIEC mice. Further, HFD enhances the number of ILC1 in the intestinal lamina propria of Sox9 ΔIEC mice and increases intestinal permeability and the accumulation of immune cells (inflammatory macrophages and T cells, and B cells) in abdominal fat tissues of obese Sox9 ΔIEC . Transplantation of fecal materials from Sox9 ΔIEC mice in germ-free mice before HFD further confirmed the regulatory role of Paneth cells for gut ILC subsets and the development of obesity.
Article
Full-text available
Diabetes mellitus (DM) is a highly prevalent disease worldwide, estimated to affect 1 in every 11 adults; among them, 90–95% of cases are type 2 diabetes mellitus. This is partly attributed to the surge in the prevalence of obesity, which has reached epidemic proportions since 2008. In these patients, cardiovascular (CV) risk stands as the primary cause of morbidity and mortality, placing a substantial burden on healthcare systems due to the potential for macrovascular and microvascular complications. In this context, leptin, an adipocyte-derived hormone, plays a fundamental role. This hormone is essential for regulating the cellular metabolism and energy balance, controlling inflammatory responses, and maintaining CV system homeostasis. Thus, leptin resistance not only contributes to weight gain but may also lead to increased cardiac inflammation, greater fibrosis, hypertension, and impairment of the cardiac metabolism. Understanding the relationship between leptin resistance and CV risk in obese individuals with type 2 DM (T2DM) could improve the management and prevention of this complication. Therefore, in this narrative review, we will discuss the evidence linking leptin with the presence, severity, and/or prognosis of obesity and T2DM regarding CV disease, aiming to shed light on the potential implications for better management and preventive strategies.
Article
Obesity is characterized by the accumulation of T cells in insulin-sensitive tissues, including the visceral adipose tissue (VAT), that can interfere with the insulin signaling pathway eventually leading to insulin resistance (IR) and type 2 diabetes. Here, we found that PD-1⁺CD4 conventional T (Tconv) cells, endowed with a transcriptomic and functional profile of partially dysfunctional cells, are diminished in VAT of obese patients with dysglycemia (OB-Dys), without a concomitant increase in apoptosis. These cells showed enhanced capacity to recirculate into the bloodstream and had a non-restricted TCRβ repertoire divergent from that of normoglycemic obese and lean individuals. PD-1⁺CD4 Tconv were reduced in the circulation of OB-Dys, exhibited an altered migration potential, and were detected in the liver of patients with non-alcoholic steatohepatitis. The findings suggest a potential role for partially dysfunctional PD-1⁺CD4 Tconv cells as inter-organ mediators of IR in obese patients with dysglycemic.
Article
Introduction Periodontitis and obesity are major health hazards. Adiposity stimulates cytokines which aggravate periodontitis. To balance this body secretes an anti-inflammatory adipokine-like visceral adipose tissue-derived serine protease inhibitor (VASPIN). Aims and Objectives The study aimed to assess serum VASPIN levels in females with and without periodontitis, categorized by obesity status. Materials and Methods Eighty female subjects with the age range of 35–44 years were included in this observational study. All have been categorized into four groups, 20 in each group. The division was based on the body mass index (BMI) and Russell’s Periodontal Index (PI). Group I-with BMI 18.5–24.9 kg/m ² ; without PD (non-obese without PD) Group II-with BMI 25-29.9 kg/m ² ; with PD (non-obese with PD). Group III -with BMI ≥30 kg/m ² ; without PD (obese without PD).Group IV with BMI ≥30 kg/m ² ; with PD (Obese with PD). Statistics Used Serum VASPIN levels were compared in all the groups. Among the groups were analyzed with ANOVA, Bartlett’s test for homogeneity of variances, and Bonferroni test for multiple comparisons. Results When VASPIN and BMI were correlated, higher levels were found in Group II, followed by Groups IV, III, and I. Serum VASPIN levels among the groups were statistically significant P < 0.0001. Conclusion Serum VASPIN can be used for predicting the progression of periodontal inflammation associated with obesity and also can act as a good biomarker for the initial stages of obesity.
Article
Full-text available
Assessment and characterization of gut microbiota has become a major research area in human disease, including type 2 diabetes, the most prevalent endocrine disease worldwide. To carry out analysis on gut microbial content in patients with type 2 diabetes, we developed a protocol for a metagenome-wide association study (MGWAS) and undertook a two-stage MGWAS based on deep shotgun sequencing of the gut microbial DNA from 345 Chinese individuals. We identified and validated approximately 60,000 type-2-diabetes-associated markers and established the concept of a metagenomic linkage group, enabling taxonomic species-level analyses. MGWAS analysis showed that patients with type 2 diabetes were characterized by a moderate degree of gut microbial dysbiosis, a decrease in the abundance of some universal butyrate-producing bacteria and an increase in various opportunistic pathogens, as well as an enrichment of other microbial functions conferring sulphate reduction and oxidative stress resistance. An analysis of 23 additional individuals demonstrated that these gut microbial markers might be useful for classifying type 2 diabetes.
Article
Full-text available
An important cause of obesity-induced insulin resistance is chronic systemic inflammation originating in visceral adipose tissue (VAT). VAT inflammation is associated with the accumulation of proinflammatory macrophages in adipose tissue, but the immunological signals that trigger their accumulation remain unknown. We found that a phenotypically distinct population of tissue-resident natural killer (NK) cells represented a crucial link between obesity-induced adipose stress and VAT inflammation. Obesity drove the upregulation of ligands of the NK cell-activating receptor NCR1 on adipocytes; this stimulated NK cell proliferation and interferon-γ (IFN-γ) production, which in turn triggered the differentiation of proinflammatory macrophages and promoted insulin resistance. Deficiency of NK cells, NCR1 or IFN-γ prevented the accumulation of proinflammatory macrophages in VAT and greatly ameliorated insulin sensitivity. Thus NK cells are key regulators of macrophage polarization and insulin resistance in response to obesity-induced adipocyte stress.
Article
Group 2 innate lymphoid cells (ILC2s) and regulatory T (Treg) cells are systemically induced by helminth infection but also sustain metabolic homeostasis in adipose tissue and contribute to tissue repair during injury. Here we show that interleukin-33 (IL-33) mediates activation of ILC2s and Treg cells in resting adipose tissue, but also after helminth infection or treatment with IL-2. Unexpectedly, ILC2-intrinsic IL-33 activation was required for Treg cell accumulation in vivo and was independent of ILC2 type 2 cytokines but partially dependent on direct co-stimulatory interactions via ICOSL-ICOS. IFN-γ inhibited ILC2 activation and Treg cell accumulation by IL-33 in infected tissue, as well as adipose tissue, where repression increased with aging and high-fat diet-induced obesity. IL-33 and ILC2s are central mediators of type 2 immune responses that promote tissue and metabolic homeostasis, and IFN-γ suppresses this pathway, likely to promote inflammatory responses and divert metabolic resources necessary to protect the host. Copyright © 2015 Elsevier Inc. All rights reserved.
Article
Interleukin-33 (IL-33) is a nuclear-associated cytokine of the IL-1 family originally described as a potent inducer of allergic type 2 immunity. IL-33 signals via the receptor ST2, which is highly expressed on group 2 innate lymphoid cells (ILC2s) and T helper 2 (Th2) cells, thus underpinning its association with helminth infection and allergic pathology. Recent studies have revealed ST2 expression on subsets of regulatory T cells, and for a role for IL-33 in tissue homeostasis and repair that suggests previously unrecognized interactions within these cellular networks. IL-33 can participate in pathologic fibrotic reactions, or, in the setting of microbial invasion, can cooperate with inflammatory cytokines to promote responses by cytotoxic NK cells, Th1 cells, and CD8(+) T cells. Here, we highlight the regulation and function of IL-33 and ST2 and review their roles in homeostasis, damage, and inflammation, suggesting a conceptual framework for future studies. Copyright © 2015 Elsevier Inc. All rights reserved.
Article
To assess the association of fasting plasma nonesterified fatty acid (NEFA) concentration with the risk of death from coronary heart disease and cancer, the authors computed 15-year mortality rates for the 4,589 working men aged 43-53 years who were included in the Paris Prospective Study between 1967 and 1972. A total of 251 and 126 men died from cancer and coronary heart disease, respectively. For coronary heart disease death, the age- and tobacco-adjusted relative risk for men in the highest 20% of the fasting plasma NEFA concentrations compared with those in the lowest 80% was 1.54 (95% confidence interval (Cl): 1.01, 2.34). It became nonsignificant after further adjustment for blood pressure, iliac/thigh ratio, and plasma insulin and cholesterol concentrations. In contrast, a high fasting plasma NEFA concentration exhibited a strong independent relation with cancer mortality (relative risk = 1.66, 95% Cl: 1.25, 2.21, after adjustment for age, cigarette consumption, heart rate, and body mass index). Despite pathophysiologic mechanisms linking NEFA metabolism with visceral fat and plasma glucose, insulin, and triglyceride concentrations, the plasma NEFA concentration does not appear to be a good marker for coronary heart disease risk. In contrast, an unexpected association with cancer mortality was found that may point to the need for further investigation.
Article
Interleukin-6 (IL-6) plays a paradoxical role in inflammation and metabolism. The pro-inflammatory effects of IL-6 are mediated via IL-6 "trans-signaling," a process where the soluble form of the IL-6 receptor (sIL-6R) binds IL-6 and activates signaling in inflammatory cells that express the gp130 but not the IL-6 receptor. Here we show that trans-signaling recruits macrophages into adipose tissue (ATM). Moreover, blocking trans-signaling with soluble gp130Fc protein prevents high-fat diet (HFD)-induced ATM accumulation, but does not improve insulin action. Importantly, however, blockade of IL-6 trans-signaling, unlike complete ablation of IL-6 signaling, does not exacerbate obesity-induced weight gain, liver steatosis, or insulin resistance. Our data identify the sIL-6R as a critical chemotactic signal for ATM recruitment and suggest that selectively blocking IL-6 trans-signaling may be a more favorable treatment option for inflammatory diseases, compared with current treatments that completely block the action of IL-6 and negatively impact upon metabolic homeostasis. Copyright © 2015 Elsevier Inc. All rights reserved.
Article
Excessive accumulation of white adipose tissue (WAT) is the defining characteristic of obesity. WAT mass is composed primarily of mature adipocytes, which are generated through the proliferation and differentiation of adipocyte precursors (APs). Although the production of new adipocytes contributes to WAT growth in obesity, little is known about the cellular and molecular mechanisms underlying adipogenesis in vivo. Here, we show that high-fat diet feeding in mice rapidly and transiently induces proliferation of APs within WAT to produce new adipocytes. Importantly, the activation of adipogenesis is specific to the perigonadal visceral depot in male mice, consistent with the patterns of obesogenic WAT growth observed in humans. Furthermore, we find that in multiple models of obesity, the activation of APs is dependent on the phosphoinositide 3-kinase (PI3K)-AKT2 pathway; however, the development of WAT does not require AKT2. These data indicate that developmental and obesogenic adipogenesis are regulated through distinct molecular mechanisms.