Sunteți pe pagina 1din 25

Accepted Manuscript

HMGB1, an innate alarmin, plays a critical role in chronic inflammation of adipose


tissue in obesity

Jing Zhang, Lei Zhang, Shu Zhang, Qilin Yu, Xiong Fei, Kun Huang, Cong-Yi Wang,
Ping Yang

PII: S0303-7207(17)30339-8
DOI: 10.1016/j.mce.2017.06.012
Reference: MCE 9984

To appear in: Molecular and Cellular Endocrinology

Received Date: 28 March 2017


Revised Date: 17 May 2017
Accepted Date: 12 June 2017

Please cite this article as: Zhang, J., Zhang, L., Zhang, S., Yu, Q., Fei, X., Huang, K., Wang, C.-Y.,
Yang, P., HMGB1, an innate alarmin, plays a critical role in chronic inflammation of adipose tissue in
obesity, Molecular and Cellular Endocrinology (2017), doi: 10.1016/j.mce.2017.06.012.

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to
our customers we are providing this early version of the manuscript. The manuscript will undergo
copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please
note that during the production process errors may be discovered which could affect the content, and all
legal disclaimers that apply to the journal pertain.
ACCEPTED MANUSCRIPT
HMGB1, an innate alarmin, plays a critical role in chronic inflammation of adipose tissue in
obesity

Jing Zhang1, Lei Zhang1, Shu Zhang1, Qilin Yu1, Xiong Fei1, Kun Huang2, Cong-Yi Wang1,*, and
Ping Yang1,*

PT
1
The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of
Education and Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University
of Science & Technology, 1095 Jiefang Ave., Wuhan, 430030, China;

RI
2
Tongji School of Pharmacy, Huazhong University of Science & Technology, Wuhan 430030,

SC
China.

*All correspondence should be addressed to Drs. Cong-Yi Wang (wangcy@tjh.tjmu.edu.cn) and

U
Ping Yang (yangping@tjh.tjmu.edu.cn), the Center for Biomedical Research, Tongji Hospital,
AN
Tongji Medical College, Huazhong University of Science & Technology, 1095 Jiefang Ave.,
Wuhan, 430030, China. Tel: 0086-27-8366-3485.
M
D
TE
C EP
AC

1
ACCEPTED MANUSCRIPT
Abstract

Obesity has emerged as an imminent global public health concern over the past several decades. It
has now become evident that obesity is characterized by the persistent and low-grade
inflammation in the adipose tissue, and serves as an independent risk factor for many metabolic
disorders such as diabetes and cardiovascular disease. Particularly, adipocytes originated from
obese mice and humans likely predominate necrosis upon stressful insults, leading to passive

PT
release of cellular contents including the high mobility group box 1 (HMGB1) into the
extracellular milieu. Extracellular HMGB1 acts as an innate alarmin to stimulate the activation of

RI
resident immune cells in the adipose tissue. Upon activation, those resident immune cells actively
secrete additional HMGB1, which in turn activates/recruits additional immune cells, and induces
adipocyte death. This review summarizes those novel discoveries in terms of HMGB1 in the

SC
initiation and maintenance of chronic inflammatory state in adipose tissue in obesity, and
discusses its potential application in clinical settings.

U
Key words: HMGB1, obesity, alarmin, DC, macrophage, adipocyte, RAGE, TLRs
AN
Abbreviations: HMGB1, high mobility group box 1; DC, dendritic cell; RAGE, receptor for
advanced glycation endproducts; TLRs, Toll-like receptors; VAT, visceral adipose tissue; WAT,
M

white adipose tissue; PAMPs, pathogen-associated molecular patterns; DAMPs,


damage-associated molecular patterns; NLS, nuclear localization sequence; SMCs, smooth muscle
cells; JAK, Janus Kinase; STAT1, Signal Transducer and Activator of Transcription; ER,
D

endoplasmic reticulum; FFA, free fatty acid; ROS, reactive oxygen species; ASCs,
TE

adipose-derived stem cells; JNK, c-Jun N-terminal kinase; NLRP3, NLR family pyrin domain
containing 3; HFD, high-fat diet; AGEs, advanced glycation endproducts; NFκB, nuclear factor
κB; T2D, type 2 diabetes; BMI, Body Mass Index
C EP
AC

2
ACCEPTED MANUSCRIPT
1. Introduction

The ongoing obesity epidemic and its associated complications of chronic diseases, exert
formidable challenges and burden to human health (Cheng et al. 2016). Obesity leads not only to
an increase in adipose tissue mass, but also to the infiltration of proinflammatory cells and
secretion of inflammatory cytokines. Therefore, obesity is characterized by a low-grade
inflammation in local and systemic sites as demonstrated by the robust secretion of

PT
proinflammatory cytokines (e.g., IL-6), and active recruitment of leukocytes (Feng et al. 2016).

HMGB1 is one of the most evolutionarily conserved proteins in the eukaryotic kingdom (Li et al.

RI
2011). It was originally identified as a chromosomal protein facilitating the binding of
transcription factors to their cognate DNA sequences (Bianchi et al. 1989). Around two decades

SC
ago, HMGB1, however, was re-discovered as an innate “danger signal” (alarmin) adopted by the
innate immune system during evolution for mediating adaptive immune responses (Wang et al.
1999; Andersson and Tracey 2004; Izuishi et al. 2006; Barkauskaite et al. 2007; Popovic et al.

U
2005). More recently, it was interestingly noted that subjects with obesity are characterized by the
AN
increase of plasma HMGB1 levels (Giacobbe et al. 2015; Arrigo et al. 2013; Wang et al. 2015),
suggesting that HMGB1 could also be a critical player for initiating and maintaining chronic
inflammatory state in adipose tissue in obesity and insulin resistance. This review intends to
M

summarize the role of HMGB1 in the initiation and maintenance of chronic inflammation state in
adipose tissue in obesity, and discuss its potential application in clinical settings.
D

2. Obesity and adipose chronic inflammation


TE

In general, visceral adipose tissue (VAT) is considered as a site of cells and molecules to control
metabolism and immune interplay relevant to energy homeostasis (Mathis 2013). In 1993,
EP

Hotamisligil and his colleagues first noted that TNF-α mRNA and protein were elevated in
adipose tissue from four different rodent models of obesity and diabetes, and blockade of TNF-α
C

in obese fa/fa rats resulted in a significant increase of peripheral glucose uptake in response to
insulin (Hotamisligil et al. 1993), suggesting that hyperglycemia could be exacerbated by cytokine
AC

overproduction. Ten years later, two groups independently reported a marked upregulation of
many inflammatory genes along with accumulation of F4/80+ macrophages in the adipose tissue
of obese mice and humans (Weisberg et al. 2003; Xu et al. 2003). These studies catalyzed the
emerging notion that chronic and low-grade inflammation is a common feature in obese
individuals, which serves as an underlying etiological factor for obesity-related chronic diseases
such as type 2 diabetes, fat-liver disease, and cardiovascular disease (Eichelmann et al. 2016).

Over past few years, our understanding of the mechanisms underlying obesity associated adipose
inflammation has been further expanded, as more cells and molecules were discovered to
3
ACCEPTED MANUSCRIPT
participate in this process. In the lean state, immune cells in adipose tissues (primarily resident
M2-like macrophages together with T regulatory (Treg) cells and eosinophils) synthesize IL-10,
IL-4, and IL-13 to maintain an anti-inflammatory environment, through which adipose tissue
maintains insulin sensitivity (Samad and Ruf 2013). In contrast, obesity drives a shift in the
number and phenotype of immune cells (Olefsky and Glass 2010; Osborn and Olefsky
2012). Specifically, monocytes are recruited from the blood into the obese adipose tissue, where
they polarize to M1 macrophages and secret proinflammatory cytokines (e.g., TNF-α, HMGB1,

PT
and IL-6) contributing to insulin resistance. Other changes accounting for the proinflammatory
state include reduced numbers of eosinophils and Tregs, and elevated numbers of neutrophils, B
cells, mast cells, and interferon γ–producing Th1 and CD8+ T cells (Wagner 2014) (Fig. 1). The

RI
proinflammatory cytokines and chemokines promote inflammation and insulin resistance acting in
autocrine, paracrine, and endocrine manners in adipose and other target tissues.

SC
Similar as the classical inflammatory responses, the initial recruitment of immune cells during

U
obesity arises from tissue derived signals in the white adipose tissue (WAT). Pathogen-associated
molecular patterns (PAMPs) are infectious and foreign “danger signals”, whereas
AN
damage-associated molecular patterns (DAMPs), for example, HMGB1, are non-infectious, yet
potentially harmful stimuli. Tissue resident immune cells express PAMP and DAMP receptors and,
upon their stimulation, initiate an inflammatory response characterized by the recruitment of more
M

immune cells in order to eradicate the stimuli by engulfing the agent or infected cells (Janeway
and Medzhitov 2002). The immune cells then initiate tissue repair and remodeling, notably by
D

clearing dead cells. The development of inflammation and dysfunction in WAT is most likely due
to a combination of endogenous and exogenous stress signals (Lumeng 2013). Indeed, obese
TE

WAT undergoes structural remodeling associated with an altered biochemical environment


(Kammoun et al. 2014). In the next several sections, we discuss with focus for the implication of
EP

HMGB1 in the pathogenesis of obesity and insulin resistance.

3. HMGB1 structural and functional properties


C
AC

HMGB1, which is abundantly expressed in almost all eukaryotic cells, was identified about 40
years ago as a nonhistone chromatin-binding protein with high electrophoretic mobility
(Andersson and Tracey 2011; Javaherian et al. 1978). The major structural features of HMGB1 are
its two DNA-binding domains termed as A and B box, and a C-terminal domain composed of
aspartic and glutamic acids (Andersson and Tracey 2011; Chen et al. 2014). HMGB1 contains two
nuclear localization sequences (NLS), which are recognized by the nuclear import complexes to
prevent its cytoplasm re-entry under physiological condition (Tang et al. 2016; Bonaldi et al. 2003;
Evankovich et al. 2010; Zhang et al. 2008; Youn and Shin 2006). Early studies demonstrated that
HMGB1 binds to the minor groove of DNA without showing sequence specificity, by which it
contributes to chromatin structure and gene expression (Liaw et al. 2016). Mice deficient in
4
ACCEPTED MANUSCRIPT
Hmgb1 die shortly after birth in part as a consequence of hypoglycemia (Calogero et al. 1999).
However, other than its nuclear role, HMGB1 has been noted to be released either passively from
damaged cells, or actively secreted by cells following cellular signal transduction (Wang et al.
1999; Tsung et al. 2007; Scaffidi et al. 2002; Gardella et al. 2002) (Fig. 2). Once in the
extracellular milieu and upon interaction with its receptors, HMGB1 exerts a plethora of functions,
including regulation of cell maturation, proliferation, migration, survival and death (Venereau et al.
2015). In addition, extracellular HMGB1 was shown to act as a chemoattractant for myeloid cells

PT
(Abraham et al. 2000), smooth muscle cells (SMCs) and meso-angioblasts, thereby promoting
muscle tissue repair (Bertheloot and Latz 2017). Interestingly, as the first critical step for its

RI
secretion from activated immune cells, its cytoplasmic translocation can be regulated by
post-translational modifications such as acetylation (Bonaldi et al. 2003), methylation and
phosphorylation (Ito et al. 2007; Oh et al. 2009; Youn and Shin 2006). For example, HMGB1 is

SC
hyperacetylated at its nuclear localization sites in monocytes and macrophages, leading to its
cytosolic relocalization. Cytosolic HMGB1 is then concentrated by default into secretory

U
lysosomes, and secreted when monocytes receive an appropriate second signal (Bonaldi et al.
2003). This process was recently recognized to be abrogated by pharmacological inhibition of the
AN
JAK/STAT1 pathway (Lu et al. 2014). However, those studies on its post-translational
modifications were conducted in a rather exclusive manner. It would be, therefore, necessary to
clarify whether its cellular localization related post-translational modification mechanisms are in
M

parallel or differentially modulated in a cell type- or cell activation-dependent manner (Bertheloot


and Latz 2017).
D

According to in vitro and in vivo studies, the immune function of HMGB1 depends on the redox
TE

forms of key cysteine residues at positions 23, 45, and 106 (Magna and Pisetsky 2016). There are
three redox forms of HMGB1, including fully reduced HMGB1, disulfide HMGB1 and sulfonyl
EP

HMGB1 (Antoine et al. 2014). Fully reduced HMGB1 without posttranslational modifications is
the predominant form in the nucleus and cytoplasm under normal conditions, and it forms a
heterocomplex with CXCL12 that interacts with receptor CXCR4 to promote cell migration
C

(Venereau et al. 2013; Lian et al. 2017). Disulfide HMGB1 with a disulfide bond connecting C23
AC

and C45 interacts with receptor TLR4 to induce cytokine and chemokine transcription and
secretion (Lian et al. 2017). Sulfonyl HMGB1 forms after exposure to ROS and has no activity for
cell migration or cytokine induction (Venereau et al. 2012). However, the investigation of specific
HMGB1 redox forms and their targeted pathways in mediating inflammation is just at its very
beginning, especially in the situation of obesity. Current and upcoming projects in this field will
be paving the way for future translational approaches targeting at the center of obesity-induced
inflammation.

4. Adipocyte necrosis and HMGB1 passive release

5
ACCEPTED MANUSCRIPT
Electron microscopy analysis revealed that in all instances, obesity-associated adipocyte death
exhibits features of necrosis, including ruptured plasma membranes, dilated endoplasmic
reticulum, cell debris in the extracellular space, and the appearance of small lipid droplets in the
cytoplasm (Ziegler and Groscurth 2004; Leist and Jaattela 2001; Imai et al. 2004; Cinti et al.
2005). These necrotic adipocytes lose their membrane integrity and release the intracellular
contents such as HMGB1 into the extracellular milieu (Wagner 2014; Rockenfeller et al. 2010),
where they act as alarmins to recruit inflammatory cells and activate resident immune cells. In

PT
sharp contrast, none of the classical ultrastructural features of apoptosis such as chromatin
condensation, plasma membrane blebbing (zeiosis), or membrane-bound apoptotic bodies with

RI
nuclear fragments, were found in obese db/db mice (Ziegler and Groscurth 2004; Leist and
Jaattela 2001; Cinti et al. 2005), suggesting that necrosis is likely to be the dominant form of
adipocyte death in obesity.

SC
Intra-abdominal pressure in obese subjects may serve as a non-pathological factor contributing to

U
adipocyte necrosis. Under physical condition, adipocytes in the abdominal cavity are generally
subject to sudden varying pressures, as occurs for example, during cough, abdominal crunch or
AN
diverse physical exercises, and in particular, obese subjects are manifested by the higher
intra-abdominal pressure (Malbrain et al. 2004; Lambert et al. 2005; Cobb et al. 2005). A visceral
adipocyte in the abdominal cavity is, thus, much more commonly exposed to rupturing forces than
M

its siblings in subcutaneous adipose tissue (Monteiro et al. 2006). As a matter of fact, adipocytes
death occurs more frequently in visceral compared with subcutaneous fat depots (Kammoun et al.
D

2014).
TE

Hypoxia in adipose tissue may serve as another driver of adipocyte death in the setting of obesity
(Kammoun et al. 2014). Hypoxia can activate stress pathways such as endoplasmic reticulum (ER)
EP

and oxidative stress with direct effects on adipocytes (Koumenis et al. 2002). Large adipocytes are
often presence in obese individuals, and their sizes in terms of diameters can reach up to 180µm,
while the diffusion distance of oxygen is only 100–200µm (Ye et al. 2007; Goossens 2008;
C

Helmlinger et al. 1997). More importantly, by employing in situ “hypoxyprobe” marker systems
AC

(such as pimonidazole) and biomarkers (e.g., leptin, plasminogen activator inhibitor type-1, matrix
metalloproteinase-2, and adrenomedullin) to measure WAT hypoxia in obese mice confirmed that
the hypoxic environment leads to devoid of nutrients, which prevents adipocytes to undergo
energy dependent apoptosis instead of necrosis (Rausch et al. 2008; Hosogai et al. 2007; Greijer
and van der Wall 2004). Furthermore, elevated concentration of free fatty acid (FFA) may serve as
a critical factor to induce adipocyte death (Rockenfeller et al. 2010; Wagner 2014). Indeed,
excessive FFA leads to mitochondria dysfunction along with accumulation of reactive oxygen
species (ROS), which then induces adipocyte necrosis. Similarly, inflammatory cytokines such as
TNF-α also induce adipocyte necrosis by increasing the production of ROS (Ventura et al. 2004).

6
ACCEPTED MANUSCRIPT
5. Active secretion of HMGB1 in WAT

Adipose tissue is vital for the life of mammals. Originally, adipose tissue was considered as an
inert depository for excess calories in the form of lipids. It has now been well recognized that
WAT also serves as an endocrine organ (Kammoun et al. 2014). In 1994, one of the first
adipocytes derived hormones, leptin, was cloned and found to be involved in the regulation of
energy homeostasis (Zhang et al. 1994). From then on, the term adipokine was used to describe

PT
any secreted substance from adipose tissue. An ever expanding number of adipokines have been
discovered including more than 50 cytokines, chemokines, hormone-like factors and other

RI
mediators (Wozniak et al. 2009; Lago et al. 2009). More recently, HMGB1 was proposed to be an
adipokine that acts as an innate pro-inflammatory mediator in WAT of patients with obesity,
which may be associated with insulin resistance and metabolic syndrome (Shimizu et al. 2016).

SC
Gunasekaran et al. demonstrated that HMGB1 was expressed 2-fold more in adipose tissue from
obese compared to normo-weight individuals, and LPS increased HMGB1 secretion by

U
adipose-derived stem cells (ASCs), but not by adipocytes isolated from human adipose tissue
(Gunasekaran et al. 2013). The human preadipocyte cell line SW872, can actively secrete HMGB1
AN
(Nativel et al. 2013). Very low level of HMGB1 release in the media could be detected in
undifferentiated 3T3-L1 preadipocytes (Shimizu et al. 2016). However, from day 4 of
differentiation, HMGB1 level in the media of 3T3-L1 adipocytes rapidly increased without any
M

stimulation. Moreover, HMGB1 is abundantly localized in the cytoplasm of 3T3-L1 adipocytes


and adipocytes of visceral adipose tissue from the obese patient (Guzman-Ruiz et al. 2014).
D

Overall, these findings indicate that HMGB1 is secreted by 3T3-L1 adipocytes, and also by the
lipid-laden adipocytes in WAT of patients with obesity. However, it is yet to be well defined by
TE

which signaling pathway HMGB1 is released by adipocytes. Inactivation of JNK in adipose tissue
decreased the production of HMGB1 by 30 percent, suggesting that apart from JNK, other
EP

signaling molecules are also involved in HMGB1 secretion from 3T3-L1 adipocytes (Shimizu et
al. 2016).
C

Other than adipocytes, a variety of other cells such as endothelial cells, preadipocytes, fibroblasts,
AC

and macrophages, are also critical components of white adipose tissue (Gesta et al. 2007; Cancello
and Clement 2006; Subramanian and Ferrante 2009). Gunasekaran et al. reported that HMGB1
production in adipose tissue mainly come from adipose-derived stromal cells (ASCs), which were
able to respond to inflammation by increasing HMGB1 secretion in a time-dependant manner
(Gunasekaran et al. 2013). Similarly, activation of endothelial inflammasomes due to increased
free fatty acids produces HMGB1, which disrupts inter-endothelial junctions and increases
paracellular permeability of endothelium contributing to early onset of endothelial injury during
obesity (Wang et al. 2016). Visfatin is another kind of adipokine, the instigation of endothelial
Nlrp3 inflammasome by visfatin and its subsequent caspase-1 activation trigger the release of
HMGB1 by endothelial cells, which acts in an autocrine or paracrine fashion to promote
7
ACCEPTED MANUSCRIPT
inter-endothelial junction disruption (Chen et al. 2015). Collectively, these results indicate that
obesity is clearly a state of low-grade chronic inflammation, and HMGB1 is definitely implicated
in this pathological process.

6. The direct effect of HMGB1 on macrophages, DCs and adipocytes

Among the immune cells that infiltrate obese adipose tissue, macrophages are functionally and

PT
numerically dominant (Ferrante 2007; Odegaard and Chawla 2011). The percentage of
macrophages in adipose tissue ranges from under 10% in lean mice and humans to over 50% in

RI
extremely obese, leptin-deficient mice and nearly 40% in obese humans, indicating that obesity
substantially alters the ratio of macrophages to adipocytes (Weisberg et al. 2003). In general,
macrophages with manifestation of a F4/80+CD11c+CD206− profile are called classically activated

SC
macrophages (M1), which can be activated either by IFN-γ or LPS to produce pro-inflammatory
cytokines (Sica and Mantovani 2012; Yao et al. 2016). While alternatively activated macrophages
(M2) are F4/80+CD11c−CD206+ and characterized by anti-inflammatory properties, and are

U
activated by IL-4 or IL-13 (Rhee 2016). It has been well established that M2 macrophages
AN
predominate in insulin-sensitive adipose tissue in lean subjects, whereas M1 macrophages
accumulate commensurate with adipose tissue mass in the obese state (Lumeng et al. 2007;
Kosteli et al. 2010; Weisberg et al. 2003). Of note, HMGB1 plays a critical role in polarizing
M

adipose-resident macrophages from M2 to an M1 phenotype (Ghosh et al. 2016). In lean subjects,


macrophages are observed interspersed among adipocytes in adipose tissue, whereas in obese
D

mice and humans, they form crown-like structures around dead or dying cells. It is noteworthy
that majority of macrophages surround necrotic adipocytes in obese mice and humans suggesting
TE

that HMGB1 released from those dysfunctional adipocytes may directly activate macrophages
(Andersson and Tracey 2011; Strissel et al. 2007; Cinti et al. 2005). In turn, adipose tissue
EP

macrophages themselves may contribute to a feed forward mechanism of adipocyte death by


releasing TNF-α and ROS (Wagner 2014).
C

Despite its importance as a professional antigen presenting cell, little is known regarding the role
AC

of dendritic cell (DC) in obese mice or humans (Cildir et al. 2013). One possible reason is that DC
and macrophage present some similar cell surface markers often leading to confusion. For
example, DC expresses CD11c which is also recognized as a marker of M1 pro-inflammatory
macrophages (Kammoun et al. 2014). However, in a recent study, Bertola et al. showed for the
first time the presence and accumulation of specific DCs in adipose tissue and that their numbers
are significantly elevated during obesity (Bertola et al. 2012). It was noted that adipose
tissue-derived HMGB1 activates TLR9 in the adipose recruited plasmacytoid DCs (pDCs) by
transporting extracellular DNA via RAGE (Ghosh et al. 2016). Extracellular HMGB1 is also
highly potent to stimulate DC activation (Messmer et al. 2004; Telusma et al. 2006; Dumitriu et al.
2005; Han et al. 2008), which then induces the expansion of CD4+ T cells as well as Th1 cells in
8
ACCEPTED MANUSCRIPT
adipose tissue, although the antigens responsible for T cell expansion are not clearly defined
(Winer et al. 2009). Th1 cells are suggested to be one of the important cells responsible for
adipose tissue inflammation and insulin resistance (Cildir et al. 2013). Consistent with this notion,
decreased Th1 cells and IFN-γ level in adipose tissue have shown to cause lower adipose tissue
inflammation and increase insulin sensitivity (Winer et al. 2009; Feuerer et al. 2009).

Unlike its effect on macrophages and DCs, the direct impact of HMGB1 on adipocyte viability

PT
and functionality is yet to be fully addressed. Generally, necrotic adipocytes lose their membrane
integrity and release HMGB1 into extracellular milieu to further impair adipocyte function,

RI
leading to obesity-associated complications such as insulin resistance and type 2 diabetes (Wagner
2014). While those discoveries are confirmative, controversial results were also reported. Feng
and colleagues reported that HMGB1 regulates autophagy in 3T3-L1 preadipocytes, and increased

SC
HMGB1 along with activated autophagy in adipose tissue of obese mice may be a protective
mechanism under stressed state induced by high-fat diet (HFD) (Feng et al. 2016). Similarly,

U
Rocío et al. reported a positive role of HMGB1 in the regulation of β-cell secretory activity
(Guzman-Ruiz et al. 2014), but this effect could turn into detrimental in response to continuous
AN
high HMGB1 levels, as occurs in obesity. Overall, additional studies would be necessary to fully
address the above observations and to dissect the underlying mechanisms.
M

7. HMGB1 signaling pathways


D

Currently, three putative signaling pathways (i.e., RAGE, TLR2 and TLR4) have been recognized
to initiate HMGB1-mediated inflammatory responses (Park et al. 2004; He et al. 2016) (Fig. 3).
TE

RAGE, a member of the immunoglobulin superfamily of cell surface molecules, has ability to
bind to and transduce the biological effects of advanced glycation endproducts (AGEs), which
EP

accumulate in settings such as hyperglycemia, aging, inflammation, renal failure, and oxidative
stress (Lopez-Diez et al. 2016; Shekhtman et al. 2017). Except for AGEs, RAGE is also a signal
transduction receptor for non-AGE molecules such as HMGB1 (Schmidt 2017). In adipose tissue
C

of obese subjects, it mainly triggers nuclear factor κB (NFκB) activation, a classical inflammatory
AC

mediator (van Beijnum et al. 2008). Of importantly note, HMGB1 binding to RAGE can
upregulate RAGE signaling, which amplifies inflammation by creating a positive feedback loop
(Yan et al. 1994). Extracellular HMGB1 also contributes to IL-6 secretion through RAGE, which
is one of the mediators that link obesity-derived chronic inflammation with insulin resistance
(Nativel et al. 2013). Particularly, adipose tissue contributes up to 35% of the circulating IL-6
(Nativel et al. 2013), which promotes low-grade inflammation to impair insulin signaling and
weaken insulin sensitivity in visceral fat (Wang et al. 2015). As a result, administration of soluble
RAGE or blocking antibody can induce a protective effect against inflammation and diabetes
(Nativel et al. 2013).

9
ACCEPTED MANUSCRIPT
Other than RAGE, TLR2 and 4 have also been characterized to be receptors for HMGB1. A great
deal of studies have consistently demonstrated that Tlr4 deficiency decreases inflammation and
insulin resistance in adipose tissue (Orr et al. 2012; Poggi et al. 2007; Tsukumo et al. 2016; Saberi
et al. 2009; Tsukumo et al. 2007; Suganami et al. 2007). Consistently, loss of Tlr4 reduces
HFD-induced hepatic steatosis and inflammation along with improved hepatic insulin sensitivity
(Orr et al. 2012; Poggi et al. 2007; Saberi et al. 2009; Tsukumo et al. 2016), and blockade of
TLR4 signaling confers protection against lipid-induced insulin resistance in skeletal muscle

PT
(Holland et al. 2011; Tsukumo et al. 2016). Similarly, TLR2 mediates FFA-induced
proinflammatory response in cultured adipocytes, macrophages, and myotubes (Konner and

RI
Bruning 2011; Nguyen et al. 2007; Senn 2006). Ablating TLR2 signaling reduces HFD-induced
lipid accumulation and inflammation in the liver and adipose tissue and generally improves
peripheral insulin sensitivity (Jin et al. 2013). Similar as RAGE signaling, ligation of HMGB1 to

SC
TLR2 or TLR4 activates NFκB, which then transcribes numerous mediators such as TNF-α, IL-6
and IL-1β to promote inflammatory response in adipose tissue (Park et al. 2003; Gunasekaran et al.

U
2016). AN
8. The feasibility of HMGB1 as an early diagnostic biomarker for obesity and therapeutic
target
M

Biomarkers are of great importance to provide valuable information for the prediction, diagnosis,
and observation of the therapeutic success of common complex multi-factorial metabolic diseases
D

such as obesity and type 2 diabetes (T2D) (Muller 2012). Based on currently available knowledge,
biomarkers used in obesity include body parameters, plasma lipids, cytokines and chemokines
TE

(Jialal et al. 2014). Diagnosis based on these traditional biomarkers, however, usually takes place
at a late stage of disease development with limited information on disease etiology (Parrizas and
EP

Novials 2016). As obesity is an important risk factor for many metabolic diseases, knowledge
about early markers may facilitate to refine prevention strategies and to provide a useful tool for
the diagnosis and treatment of obesity and its complications. Therefore, only those biomarkers
C

readily identified from easily accessible sources (e.g., blood) and associated with disease etiology
AC

could meet the demand. In this regard, serum level of HMGB1 emerges as an untapped promise.

Indeed, clinical studies demonstrated a positive correlation between HMGB1 and obesity in
pregnant women, children, and Chinese subjects (Giacobbe et al. 2015; Arrigo et al. 2013; Wang
et al. 2015). Giacobbe and colleagues evaluated serum HMGB1 levels in obese pregnant women,
and identified a direct and statistically significant correlation between BMI of the study group and
HMGB1 levels (Giacobbe et al. 2015). Similarly, Arrigo et al. reported a positive correlation
between serum HMGB1 and obesity in children (Arrigo et al. 2013). Independently, circulating
HMGB1 was noted to be significantly increased not only in obese individuals, but also in T2D
patients (Wang et al. 2015). However, these studies are manifested by some limitations such as
10
ACCEPTED MANUSCRIPT
small example size and the lack of different ethnicities. Therefore, larger sample size coupled with
different populations should be considered in the future study to further confirm the feasibility of
HMGB1 as a viable biomarker for early diagnosis of obesity.

Since HMGB1 plays a critical role in obesity-induced inflammation, antagonizing HMGB1


inflammatory pathway could bring on new therapeutic targets to counteract obesity-associated
pathologies. For example, neutralization of HMGB1 appears to reduce weight gain and liver

PT
inflammation in mice fed an obesogenic diet (Montes et al. 2015). Treating apoE-deficient mice
with anti-HMGB1 neutralizing antibody results in decreased lesions, as well as a reduction in
many inflammatory cell types such as dendritic cells, macrophages and CD4+ T cells in

RI
atherosclerotic lesions (Kanellakis et al. 2011). Metformin, a biguanide derivative, is the first-line
drug in the treatment of type 2 diabetes (Foretz et al. 2014). While recently, Horiuchi and
colleagues, identified HMGB1 as a novel metformin-binding protein and demonstrated that

SC
metformin inhibited cytokine activity of HMGB1 in vitro and in vivo (Horiuchi et al. 2017). This
is the first report of an extracellular function of metformin that regulates inflammation induced by
HMGB1, which suggests possible new ways to manage HMGB1-induced inflammation in obesity.

9. Conclusion remarks
U
AN
Obesity has emerged as an imminent global public health concern over the past several decades. It
M

is featured by the persistent and low-grade inflammation, and serves as a causative factor for many
secondary diseases including T2D, cardiovascular disease, high cholesterol, and infertility. As an
innate alarmin once released from necrotic adipocytes, HMGB1 stimulates the activation of
D

resident immune cells in the adipose tissue. Upon activation, those resident immune cells actively
TE

secrete additional HMGB1, which then activates/recruits additional immune cells, and acts on
adipocytes in autocrine, paracrine or systemic fashion. Therefore, HMGB1 plays a critical role in
the initiation and maintenance of chronic inflammatory state in adipose tissue of obese subjects.
EP

There is also feasible evidence that HMGB1 could be a viable biomarker for early diagnosis of
obesity and therapeutic target for prevention and intervention of obesity-induced inflammation.
However, additional studies would be necessary to warrant its application in clinical settings.
C
AC

Acknowledgements
Our work was supported by the National Natural Science Foundation of China (81530024,
81471046, 81470988, 81500637 and 81670729), the Program Project for Type 1 Diabetes from
the Ministry of Science and Technology (2016YFC1305002), the Integrated Innovative Team for
Major Human Disease Programs of Tongji Medical College, Huazhong University of Science &
Technology, and the Innovative Funding for Translational Research from Tongji Hospital.

11
ACCEPTED MANUSCRIPT
References
Abraham E., Arcaroli, J., Carmody, A., Wang, H.andTracey, K.J., 2000, Hmg-1 as a mediator of acute
lung inflammation. Journal of immunology 165, 2950-2954.
Andersson U.andTracey, K.J., 2004, Hmgb1 as a mediator of necrosis-induced inflammation and a
therapeutic target in arthritis. Rheumatic diseases clinics of North America 30, 627-637, xi.
Andersson U.andTracey, K.J., 2011, Hmgb1 is a therapeutic target for sterile inflammation and
infection. Annual review of immunology 29, 139-162.
Antoine D.J., Harris, H.E., Andersson, U., Tracey, K.J.andBianchi, M.E., 2014, A systematic

PT
nomenclature for the redox states of high mobility group box (hmgb) proteins. Molecular
medicine 20, 135-137.
Arrigo T., Chirico, V., Salpietro, V., Munafo, C., Ferrau, V., Gitto, E., Lacquaniti, A.andSalpietro, C.,

RI
2013, High-mobility group protein b1: A new biomarker of metabolic syndrome in obese
children. European journal of endocrinology 168, 631-638.

SC
Barkauskaite V., Ek, M., Popovic, K., Harris, H.E., Wahren-Herlenius, M.andNyberg, F., 2007,
Translocation of the novel cytokine hmgb1 to the cytoplasm and extracellular space coincides
with the peak of clinical activity in experimentally uv-induced lesions of cutaneous lupus
erythematosus. Lupus 16, 794-802.

U
Bertheloot D.andLatz, E., 2017, Hmgb1, il-1alpha, il-33 and s100 proteins: Dual-function alarmins.
AN
Cellular & molecular immunology 14, 43-64.
Bertola A., Ciucci, T., Rousseau, D., Bourlier, V., Duffaut, C., Bonnafous, S., Blin-Wakkach, C., Anty,
R., Iannelli, A., Gugenheim, J., Tran, A., Bouloumie, A., Gual, P.andWakkach, A., 2012,
M

Identification of adipose tissue dendritic cells correlated with obesity-associated


insulin-resistance and inducing th17 responses in mice and patients. Diabetes 61, 2238-2247.
Bianchi M.E., Beltrame, M.andPaonessa, G., 1989, Specific recognition of cruciform DNA by nuclear
D

protein hmg1. Science 243, 1056-1059.


Bonaldi T., Talamo, F., Scaffidi, P., Ferrera, D., Porto, A., Bachi, A., Rubartelli, A., Agresti,
TE

A.andBianchi, M.E., 2003, Monocytic cells hyperacetylate chromatin protein hmgb1 to


redirect it towards secretion. The EMBO journal 22, 5551-5560.
Calogero S., Grassi, F., Aguzzi, A., Voigtlander, T., Ferrier, P., Ferrari, S.andBianchi, M.E., 1999, The
EP

lack of chromosomal protein hmg1 does not disrupt cell growth but causes lethal
hypoglycaemia in newborn mice. Nature genetics 22, 276-280.
Cancello R.andClement, K., 2006, Is obesity an inflammatory illness? Role of low-grade inflammation
C

and macrophage infiltration in human white adipose tissue. BJOG : an international journal
of obstetrics and gynaecology 113, 1141-1147.
AC

Chen M., Huang, W., Wang, C., Nie, H., Li, G., Sun, T., Yang, F., Zhang, Y., Shu, K., Wang,
C.andGong, Q., 2014, High-mobility group box 1 exacerbates ccl(4)-induced acute liver
injury in mice. Clinical immunology 153, 56-63.
Chen Y., Pitzer, A.L., Li, X., Li, P.L., Wang, L.andZhang, Y., 2015, Instigation of endothelial nlrp3
inflammasome by adipokine visfatin promotes inter-endothelial junction disruption: Role of
hmgb1. Journal of cellular and molecular medicine 19, 2715-2727.
Cheng J., Song, J., He, X., Zhang, M., Hu, S., Zhang, S., Yu, Q., Yang, P., Xiong, F., Wang, D.W., Zhou,
J., Ning, Q., Chen, Z., Eizirik, D.L., Zhou, Z., Zhao, C.andWang, C.Y., 2016, Loss of mbd2
protects mice against high-fat diet-induced obesity and insulin resistance by regulating the
homeostasis of energy storage and expenditure. Diabetes 65, 3384-3395.

12
ACCEPTED MANUSCRIPT
Cildir G., Akincilar, S.C.andTergaonkar, V., 2013, Chronic adipose tissue inflammation: All immune
cells on the stage. Trends in molecular medicine 19, 487-500.
Cinti S., Mitchell, G., Barbatelli, G., Murano, I., Ceresi, E., Faloia, E., Wang, S., Fortier, M., Greenberg,
A.S.andObin, M.S., 2005, Adipocyte death defines macrophage localization and function in
adipose tissue of obese mice and humans. Journal of lipid research 46, 2347-2355.
Cobb W.S., Burns, J.M., Kercher, K.W., Matthews, B.D., James Norton, H.andTodd Heniford, B., 2005,
Normal intraabdominal pressure in healthy adults. The Journal of surgical research 129,
231-235.

PT
Dumitriu I.E., Baruah, P., Valentinis, B., Voll, R.E., Herrmann, M., Nawroth, P.P., Arnold, B., Bianchi,
M.E., Manfredi, A.A.andRovere-Querini, P., 2005, Release of high mobility group box 1 by
dendritic cells controls t cell activation via the receptor for advanced glycation end products.

RI
Journal of immunology 174, 7506-7515.
Eichelmann F., Schwingshackl, L., Fedirko, V.andAleksandrova, K., 2016, Effect of plant-based diets

SC
on obesity-related inflammatory profiles: A systematic review and meta-analysis of
intervention trials. Obesity reviews : an official journal of the International Association for the
Study of Obesity 17, 1067-1079.
Evankovich J., Cho, S.W., Zhang, R., Cardinal, J., Dhupar, R., Zhang, L., Klune, J.R., Zlotnicki, J.,

U
Billiar, T.andTsung, A., 2010, High mobility group box 1 release from hepatocytes during
AN
ischemia and reperfusion injury is mediated by decreased histone deacetylase activity. The
Journal of biological chemistry 285, 39888-39897.
Feng H., Yu, L., Zhang, G., Liu, G., Yang, C., Wang, H.andSong, X., 2016, Regulation of
autophagy-related protein and cell differentiation by high mobility group box 1 protein in
M

adipocytes. Mediators of inflammation 2016, 1936386.


Ferrante A.W., Jr., 2007, Obesity-induced inflammation: A metabolic dialogue in the language of
D

inflammation. Journal of internal medicine 262, 408-414.


Feuerer M., Herrero, L., Cipolletta, D., Naaz, A., Wong, J., Nayer, A., Lee, J., Goldfine, A.B., Benoist,
TE

C., Shoelson, S.andMathis, D., 2009, Lean, but not obese, fat is enriched for a unique
population of regulatory t cells that affect metabolic parameters. Nature medicine 15, 930-939.
Foretz M., Guigas, B., Bertrand, L., Pollak, M.andViollet, B., 2014, Metformin: From mechanisms of
EP

action to therapies. Cell metabolism 20, 953-966.


Gardella S., Andrei, C., Ferrera, D., Lotti, L.V., Torrisi, M.R., Bianchi, M.E.andRubartelli, A., 2002,
The nuclear protein hmgb1 is secreted by monocytes via a non-classical, vesicle-mediated
C

secretory pathway. EMBO reports 3, 995-1001.


Gesta S., Tseng, Y.H.andKahn, C.R., 2007, Developmental origin of fat: Tracking obesity to its source.
AC

Cell 131, 242-256.


Ghosh A.R., Bhattacharya, R., Bhattacharya, S., Nargis, T., Rahaman, O., Duttagupta, P., Raychaudhuri,
D., Liu, C.S., Roy, S., Ghosh, P., Khanna, S., Chaudhuri, T., Tantia, O., Haak, S.,
Bandyopadhyay, S., Mukhopadhyay, S., Chakrabarti, P.andGanguly, D., 2016, Adipose
recruitment and activation of plasmacytoid dendritic cells fuel metaflammation. Diabetes 65,
3440-3452.
Giacobbe A., Grasso, R., Imbesi, G., Salpietro, C.D., Grasso, L., Lagana, A.S., Triolo, O.andDi
Benedetto, A., 2015, High mobility group protein b1: A new biomarker of obesity in pregnant
women? Gynecological endocrinology : the official journal of the International Society of
Gynecological Endocrinology 31, 113-115.

13
ACCEPTED MANUSCRIPT
Goossens G.H., 2008, The role of adipose tissue dysfunction in the pathogenesis of obesity-related
insulin resistance. Physiology & behavior 94, 206-218.
Greijer A.E.andvan der Wall, E., 2004, The role of hypoxia inducible factor 1 (hif-1) in hypoxia
induced apoptosis. Journal of clinical pathology 57, 1009-1014.
Gunasekaran M.K., Virama-Latchoumy, A.L., Girard, A.C., Planesse, C., Guerin-Dubourg, A., Ottosson,
L., Andersson, U., Cesari, M., Roche, R.andHoareau, L., 2016, Tlr4-dependant
pro-inflammatory effects of hmgb1 on human adipocyte. Adipocyte 5, 384-388.
Gunasekaran M.K., Viranaicken, W., Girard, A.C., Festy, F., Cesari, M., Roche, R.andHoareau, L.,

PT
2013, Inflammation triggers high mobility group box 1 (hmgb1) secretion in adipose tissue, a
potential link to obesity. Cytokine 64, 103-111.
Guzman-Ruiz R., Ortega, F., Rodriguez, A., Vazquez-Martinez, R., Diaz-Ruiz, A., Garcia-Navarro, S.,

RI
Giralt, M., Garcia-Rios, A., Cobo-Padilla, D., Tinahones, F.J., Lopez-Miranda, J., Villarroya,
F., Fruhbeck, G., Fernandez-Real, J.M.andMalagon, M.M., 2014, Alarmin high-mobility

SC
group b1 (hmgb1) is regulated in human adipocytes in insulin resistance and influences insulin
secretion in beta-cells. International journal of obesity 38, 1545-1554.
Han J., Zhong, J., Wei, W., Wang, Y., Huang, Y., Yang, P., Purohit, S., Dong, Z., Wang, M.H., She, J.X.,
Gong, F., Stern, D.M.andWang, C.Y., 2008, Extracellular high-mobility group box 1 acts as an

U
innate immune mediator to enhance autoimmune progression and diabetes onset in nod mice.
AN
Diabetes 57, 2118-2127.
He L., Sun, F., Wang, Y., Zhu, J., Fang, J., Zhang, S., Yu, Q., Gong, Q., Ren, B., Xiang, X., Chen, Z.,
Ning, Q., Hu, J., Yang, P.andWang, C.Y., 2016, Hmgb1 exacerbates bronchiolitis obliterans
syndrome via rage/nf-kappab/hpse signaling to enhance latent tgf-beta release from ecm.
M

American journal of translational research 8, 1971-1984.


Helmlinger G., Yuan, F., Dellian, M.andJain, R.K., 1997, Interstitial ph and po2 gradients in solid
D

tumors in vivo: High-resolution measurements reveal a lack of correlation. Nature medicine 3,


177-182.
TE

Holland W.L., Bikman, B.T., Wang, L.P., Yuguang, G., Sargent, K.M., Bulchand, S., Knotts, T.A., Shui,
G., Clegg, D.J., Wenk, M.R., Pagliassotti, M.J., Scherer, P.E.andSummers, S.A., 2011,
Lipid-induced insulin resistance mediated by the proinflammatory receptor tlr4 requires
EP

saturated fatty acid-induced ceramide biosynthesis in mice. The Journal of clinical


investigation 121, 1858-1870.
Horiuchi T., Sakata, N., Narumi, Y., Kimura, T., Hayashi, T., Nagano, K., Liu, K., Nishibori, M.,
C

Tsukita, S., Yamada, T., Katagiri, H., Shirakawa, R.andHoriuchi, H., 2017, Metformin directly
binds the alarmin hmgb1 and inhibits its proinflammatory activity. The Journal of biological
AC

chemistry.
Hosogai N., Fukuhara, A., Oshima, K., Miyata, Y., Tanaka, S., Segawa, K., Furukawa, S., Tochino, Y.,
Komuro, R., Matsuda, M.andShimomura, I., 2007, Adipose tissue hypoxia in obesity and its
impact on adipocytokine dysregulation. Diabetes 56, 901-911.
Hotamisligil G.S., Shargill, N.S.andSpiegelman, B.M., 1993, Adipose expression of tumor necrosis
factor-alpha: Direct role in obesity-linked insulin resistance. Science 259, 87-91.
Imai T., Takakuwa, R., Marchand, S., Dentz, E., Bornert, J.M., Messaddeq, N., Wendling, O., Mark, M.,
Desvergne, B., Wahli, W., Chambon, P.andMetzger, D., 2004, Peroxisome
proliferator-activated receptor gamma is required in mature white and brown adipocytes for
their survival in the mouse. Proceedings of the National Academy of Sciences of the United

14
ACCEPTED MANUSCRIPT
States of America 101, 4543-4547.
Ito I., Fukazawa, J.andYoshida, M., 2007, Post-translational methylation of high mobility group box 1
(hmgb1) causes its cytoplasmic localization in neutrophils. The Journal of biological
chemistry 282, 16336-16344.
Izuishi K., Tsung, A., Jeyabalan, G., Critchlow, N.D., Li, J., Tracey, K.J., Demarco, R.A., Lotze, M.T.,
Fink, M.P., Geller, D.A.andBilliar, T.R., 2006, Cutting edge: High-mobility group box 1
preconditioning protects against liver ischemia-reperfusion injury. Journal of immunology 176,
7154-7158.

PT
Janeway C.A., Jr.andMedzhitov, R., 2002, Innate immune recognition. Annual review of immunology
20, 197-216.
Javaherian K., Liu, J.F.andWang, J.C., 1978, Nonhistone proteins hmg1 and hmg2 change the DNA

RI
helical structure. Science 199, 1345-1346.
Jialal I., Kaur, H.andDevaraj, S., 2014, Toll-like receptor status in obesity and metabolic syndrome: A

SC
translational perspective. The Journal of clinical endocrinology and metabolism 99, 39-48.
Jin C., Henao-Mejia, J.andFlavell, R.A., 2013, Innate immune receptors: Key regulators of metabolic
disease progression. Cell metabolism 17, 873-882.
Kammoun H.L., Kraakman, M.J.andFebbraio, M.A., 2014, Adipose tissue inflammation in glucose

U
metabolism. Reviews in endocrine & metabolic disorders 15, 31-44.
AN
Kanellakis P., Agrotis, A., Kyaw, T.S., Koulis, C., Ahrens, I., Mori, S., Takahashi, H.K., Liu, K., Peter,
K., Nishibori, M.andBobik, A., 2011, High-mobility group box protein 1 neutralization
reduces development of diet-induced atherosclerosis in apolipoprotein e-deficient mice.
Arteriosclerosis, thrombosis, and vascular biology 31, 313-319.
M

Konner A.C.andBruning, J.C., 2011, Toll-like receptors: Linking inflammation to metabolism. Trends
in endocrinology and metabolism: TEM 22, 16-23.
D

Kosteli A., Sugaru, E., Haemmerle, G., Martin, J.F., Lei, J., Zechner, R.andFerrante, A.W., Jr., 2010,
Weight loss and lipolysis promote a dynamic immune response in murine adipose tissue. The
TE

Journal of clinical investigation 120, 3466-3479.


Koumenis C., Naczki, C., Koritzinsky, M., Rastani, S., Diehl, A., Sonenberg, N., Koromilas,
A.andWouters, B.G., 2002, Regulation of protein synthesis by hypoxia via activation of the
EP

endoplasmic reticulum kinase perk and phosphorylation of the translation initiation factor
eif2alpha. Molecular and cellular biology 22, 7405-7416.
Lago F., Gomez, R., Gomez-Reino, J.J., Dieguez, C.andGualillo, O., 2009, Adipokines as novel
C

modulators of lipid metabolism. Trends in biochemical sciences 34, 500-510.


Lambert D.M., Marceau, S.andForse, R.A., 2005, Intra-abdominal pressure in the morbidly obese.
AC

Obesity surgery 15, 1225-1232.


Leist M.andJaattela, M., 2001, Four deaths and a funeral: From caspases to alternative mechanisms.
Nature reviews. Molecular cell biology 2, 589-598.
Li J., Gong, Q., Zhong, S., Wang, L., Guo, H., Xiang, Y., Ichim, T.E., Wang, C.Y., Chen, S., Gong,
F.andChen, G., 2011, Neutralization of the extracellular hmgb1 released by ischaemic
damaged renal cells protects against renal ischaemia-reperfusion injury. Nephrology, dialysis,
transplantation : official publication of the European Dialysis and Transplant Association -
European Renal Association 26, 469-478.
Lian Y.J., Gong, H., Wu, T.Y., Su, W.J., Zhang, Y., Yang, Y.Y., Peng, W., Zhang, T., Zhou, J.R., Jiang,
C.L.andWang, Y.X., 2017, Ds-hmgb1 and fr-hmgb induce depressive behavior through

15
ACCEPTED MANUSCRIPT
neuroinflammation in contrast to nonoxid-hmgb1. Brain, behavior, and immunity 59, 322-332.
Liaw P.C., Ito, T., Iba, T., Thachil, J.andZeerleder, S., 2016, Damp and dic: The role of extracellular
DNA and DNA-binding proteins in the pathogenesis of dic. Blood reviews 30, 257-261.
Lopez-Diez R., Shekhtman, A., Ramasamy, R.andSchmidt, A.M., 2016, Cellular mechanisms and
consequences of glycation in atherosclerosis and obesity. Biochimica et biophysica acta 1862,
2244-2252.
Lu B., Antoine, D.J., Kwan, K., Lundback, P., Wahamaa, H., Schierbeck, H., Robinson, M., Van Zoelen,
M.A., Yang, H., Li, J., Erlandsson-Harris, H., Chavan, S.S., Wang, H., Andersson,

PT
U.andTracey, K.J., 2014, Jak/stat1 signaling promotes hmgb1 hyperacetylation and nuclear
translocation. Proceedings of the National Academy of Sciences of the United States of
America 111, 3068-3073.

RI
Lumeng C.N., 2013, Innate immune activation in obesity. Molecular aspects of medicine 34, 12-29.
Lumeng C.N., Bodzin, J.L.andSaltiel, A.R., 2007, Obesity induces a phenotypic switch in adipose

SC
tissue macrophage polarization. The Journal of clinical investigation 117, 175-184.
Magna M.andPisetsky, D.S., 2016, The alarmin properties of DNA and DNA-associated nuclear
proteins. Clinical therapeutics 38, 1029-1041.
Malbrain M.L., Chiumello, D., Pelosi, P., Wilmer, A., Brienza, N., Malcangi, V., Bihari, D., Innes, R.,

U
Cohen, J., Singer, P., Japiassu, A., Kurtop, E., De Keulenaer, B.L., Daelemans, R., Del Turco,
AN
M., Cosimini, P., Ranieri, M., Jacquet, L., Laterre, P.F.andGattinoni, L., 2004, Prevalence of
intra-abdominal hypertension in critically ill patients: A multicentre epidemiological study.
Intensive care medicine 30, 822-829.
Mathis D., 2013, Immunological goings-on in visceral adipose tissue. Cell metabolism 17, 851-859.
M

Messmer D., Yang, H., Telusma, G., Knoll, F., Li, J., Messmer, B., Tracey, K.J.andChiorazzi, N., 2004,
High mobility group box protein 1: An endogenous signal for dendritic cell maturation and th1
D

polarization. Journal of immunology 173, 307-313.


Monteiro R., de Castro, P.M., Calhau, C.andAzevedo, I., 2006, Adipocyte size and liability to cell death.
TE

Obesity surgery 16, 804-806.


Montes V.N., Subramanian, S., Goodspeed, L., Wang, S.A., Omer, M., Bobik, A., Teshigawara, K.,
Nishibori, M.andChait, A., 2015, Anti-hmgb1 antibody reduces weight gain in mice fed a
EP

high-fat diet. Nutrition & diabetes 5, e161.


Muller G., 2012, Microvesicles/exosomes as potential novel biomarkers of metabolic diseases.
Diabetes, metabolic syndrome and obesity : targets and therapy 5, 247-282.
C

Nativel B., Marimoutou, M., Thon-Hon, V.G., Gunasekaran, M.K., Andries, J., Stanislas, G., Planesse,
C., Da Silva, C.R., Cesari, M., Iwema, T., Gasque, P.andViranaicken, W., 2013, Soluble
AC

hmgb1 is a novel adipokine stimulating il-6 secretion through rage receptor in sw872
preadipocyte cell line: Contribution to chronic inflammation in fat tissue. PloS one 8, e76039.
Nguyen M.T., Favelyukis, S., Nguyen, A.K., Reichart, D., Scott, P.A., Jenn, A., Liu-Bryan, R., Glass,
C.K., Neels, J.G.andOlefsky, J.M., 2007, A subpopulation of macrophages infiltrates
hypertrophic adipose tissue and is activated by free fatty acids via toll-like receptors 2 and 4
and jnk-dependent pathways. The Journal of biological chemistry 282, 35279-35292.
Odegaard J.I.andChawla, A., 2011, Alternative macrophage activation and metabolism. Annual review
of pathology 6, 275-297.
Oh Y.J., Youn, J.H., Ji, Y., Lee, S.E., Lim, K.J., Choi, J.E.andShin, J.S., 2009, Hmgb1 is
phosphorylated by classical protein kinase c and is secreted by a calcium-dependent

16
ACCEPTED MANUSCRIPT
mechanism. Journal of immunology 182, 5800-5809.
Olefsky J.M.andGlass, C.K., 2010, Macrophages, inflammation, and insulin resistance. Annual review
of physiology 72, 219-246.
Orr J.S., Puglisi, M.J., Ellacott, K.L., Lumeng, C.N., Wasserman, D.H.andHasty, A.H., 2012, Toll-like
receptor 4 deficiency promotes the alternative activation of adipose tissue macrophages.
Diabetes 61, 2718-2727.
Osborn O.andOlefsky, J.M., 2012, The cellular and signaling networks linking the immune system and
metabolism in disease. Nature medicine 18, 363-374.

PT
Park J.S., Arcaroli, J., Yum, H.K., Yang, H., Wang, H., Yang, K.Y., Choe, K.H., Strassheim, D., Pitts,
T.M., Tracey, K.J.andAbraham, E., 2003, Activation of gene expression in human neutrophils
by high mobility group box 1 protein. American journal of physiology. Cell physiology 284,

RI
C870-879.
Park J.S., Svetkauskaite, D., He, Q., Kim, J.Y., Strassheim, D., Ishizaka, A.andAbraham, E., 2004,

SC
Involvement of toll-like receptors 2 and 4 in cellular activation by high mobility group box 1
protein. The Journal of biological chemistry 279, 7370-7377.
Parrizas M.andNovials, A., 2016, Circulating micrornas as biomarkers for metabolic disease. Best
practice & research. Clinical endocrinology & metabolism 30, 591-601.

U
Poggi M., Bastelica, D., Gual, P., Iglesias, M.A., Gremeaux, T., Knauf, C., Peiretti, F., Verdier, M.,
AN
Juhan-Vague, I., Tanti, J.F., Burcelin, R.andAlessi, M.C., 2007, C3h/hej mice carrying a
toll-like receptor 4 mutation are protected against the development of insulin resistance in
white adipose tissue in response to a high-fat diet. Diabetologia 50, 1267-1276.
Popovic K., Ek, M., Espinosa, A., Padyukov, L., Harris, H.E., Wahren-Herlenius, M.andNyberg, F.,
M

2005, Increased expression of the novel proinflammatory cytokine high mobility group box
chromosomal protein 1 in skin lesions of patients with lupus erythematosus. Arthritis and
D

rheumatism 52, 3639-3645.


Rausch M.E., Weisberg, S., Vardhana, P.andTortoriello, D.V., 2008, Obesity in c57bl/6j mice is
TE

characterized by adipose tissue hypoxia and cytotoxic t-cell infiltration. International journal
of obesity 32, 451-463.
Rhee I., 2016, Diverse macrophages polarization in tumor microenvironment. Archives of pharmacal
EP

research 39, 1588-1596.


Rockenfeller P., Ring, J., Muschett, V., Beranek, A., Buettner, S., Carmona-Gutierrez, D., Eisenberg, T.,
Khoury, C., Rechberger, G., Kohlwein, S.D., Kroemer, G.andMadeo, F., 2010, Fatty acids
C

trigger mitochondrion-dependent necrosis. Cell cycle 9, 2836-2842.


Saberi M., Woods, N.B., de Luca, C., Schenk, S., Lu, J.C., Bandyopadhyay, G., Verma, I.M.andOlefsky,
AC

J.M., 2009, Hematopoietic cell-specific deletion of toll-like receptor 4 ameliorates hepatic and
adipose tissue insulin resistance in high-fat-fed mice. Cell metabolism 10, 419-429.
Samad F.andRuf, W., 2013, Inflammation, obesity, and thrombosis. Blood 122, 3415-3422.
Scaffidi P., Misteli, T.andBianchi, M.E., 2002, Release of chromatin protein hmgb1 by necrotic cells
triggers inflammation. Nature 418, 191-195.
Schmidt A.M., 2017, 2016 atvb plenary lecture: Receptor for advanced glycation endproducts and
implications for the pathogenesis an treatment of cardiometabolic disorders: Spotlight on the
macrophage. Arteriosclerosis, thrombosis, and vascular biology.
Senn J.J., 2006, Toll-like receptor-2 is essential for the development of palmitate-induced insulin
resistance in myotubes. The Journal of biological chemistry 281, 26865-26875.

17
ACCEPTED MANUSCRIPT
Shekhtman A., Ramasamy, R.andSchmidt, A.M., 2017, Glycation & the rage axis: Targeting signal
transduction through diaph1. Expert review of proteomics 14, 147-156.
Shimizu T., Yamakuchi, M., Biswas, K.K., Aryal, B., Yamada, S., Hashiguchi, T.andMaruyama, I.,
2016, Hmgb1 is secreted by 3t3-l1 adipocytes through jnk signaling and the secretion is
partially inhibited by adiponectin. Obesity 24, 1913-1921.
Sica A.andMantovani, A., 2012, Macrophage plasticity and polarization: In vivo veritas. The Journal of
clinical investigation 122, 787-795.
Strissel K.J., Stancheva, Z., Miyoshi, H., Perfield, J.W., 2nd, DeFuria, J., Jick, Z., Greenberg,

PT
A.S.andObin, M.S., 2007, Adipocyte death, adipose tissue remodeling, and obesity
complications. Diabetes 56, 2910-2918.
Subramanian V.andFerrante, A.W., Jr., 2009, Obesity, inflammation, and macrophages. Nestle Nutrition

RI
workshop series. Paediatric programme 63, 151-159; discussion 159-162, 259-168.
Suganami T., Mieda, T., Itoh, M., Shimoda, Y., Kamei, Y.andOgawa, Y., 2007, Attenuation of

SC
obesity-induced adipose tissue inflammation in c3h/hej mice carrying a toll-like receptor 4
mutation. Biochemical and biophysical research communications 354, 45-49.
Tang Y., Zhao, X., Antoine, D., Xiao, X., Wang, H., Andersson, U., Billiar, T.R., Tracey, K.J.andLu, B.,
2016, Regulation of posttranslational modifications of hmgb1 during immune responses.

U
Antioxidants & redox signaling 24, 620-634.
AN
Telusma G., Datta, S., Mihajlov, I., Ma, W., Li, J., Yang, H., Newman, W., Messmer, B.T., Minev, B.,
Schmidt-Wolf, I.G., Tracey, K.J., Chiorazzi, N.andMessmer, D., 2006, Dendritic cell
activating peptides induce distinct cytokine profiles. International immunology 18,
1563-1573.
M

Tsukumo D.M., Carvalho-Filho, M.A., Carvalheira, J.B., Prada, P.O., Hirabara, S.M., Schenka, A.A.,
Araujo, E.P., Vassallo, J., Curi, R., Velloso, L.A.andSaad, M.J., 2007, Loss-of-function
D

mutation in toll-like receptor 4 prevents diet-induced obesity and insulin resistance. Diabetes
56, 1986-1998.
TE

Tsukumo D.M., Carvalho-Filho, M.A., Carvalheira, J.B., Prada, P.O., Hirabara, S.M., Schenka, A.A.,
Araujo, E.P., Vassallo, J., Curi, R., Velloso, L.A.andSaad, M.J., 2016, Statement of retraction.
Loss-of-function mutation in toll-like receptor 4 prevents diet-induced obesity and insulin
EP

resistance. Diabetes 2007;56:1986-1998. Doi: 10.2337/db06-1595. Diabetes 65, 1126-1127.


Tsung A., Klune, J.R., Zhang, X., Jeyabalan, G., Cao, Z., Peng, X., Stolz, D.B., Geller, D.A., Rosengart,
M.R.andBilliar, T.R., 2007, Hmgb1 release induced by liver ischemia involves toll-like
C

receptor 4 dependent reactive oxygen species production and calcium-mediated signaling. The
Journal of experimental medicine 204, 2913-2923.
AC

van Beijnum J.R., Buurman, W.A.andGriffioen, A.W., 2008, Convergence and amplification of toll-like
receptor (tlr) and receptor for advanced glycation end products (rage) signaling pathways via
high mobility group b1 (hmgb1). Angiogenesis 11, 91-99.
Venereau E., Casalgrandi, M., Schiraldi, M., Antoine, D.J., Cattaneo, A., De Marchis, F., Liu, J.,
Antonelli, A., Preti, A., Raeli, L., Shams, S.S., Yang, H., Varani, L., Andersson, U., Tracey,
K.J., Bachi, A., Uguccioni, M.andBianchi, M.E., 2012, Mutually exclusive redox forms of
hmgb1 promote cell recruitment or proinflammatory cytokine release. The Journal of
experimental medicine 209, 1519-1528.
Venereau E., Ceriotti, C.andBianchi, M.E., 2015, Damps from cell death to new life. Frontiers in
immunology 6, 422.

18
ACCEPTED MANUSCRIPT
Venereau E., Schiraldi, M., Uguccioni, M.andBianchi, M.E., 2013, Hmgb1 and leukocyte migration
during trauma and sterile inflammation. Molecular immunology 55, 76-82.
Ventura J.J., Cogswell, P., Flavell, R.A., Baldwin, A.S., Jr.andDavis, R.J., 2004, Jnk potentiates
tnf-stimulated necrosis by increasing the production of cytotoxic reactive oxygen species.
Genes & development 18, 2905-2915.
Wagner M., 2014, A dangerous duo in adipose tissue: High-mobility group box 1 protein and
macrophages. The Yale journal of biology and medicine 87, 127-133.
Wang H., Bloom, O., Zhang, M., Vishnubhakat, J.M., Ombrellino, M., Che, J., Frazier, A., Yang, H.,

PT
Ivanova, S., Borovikova, L., Manogue, K.R., Faist, E., Abraham, E., Andersson, J., Andersson,
U., Molina, P.E., Abumrad, N.N., Sama, A.andTracey, K.J., 1999, Hmg-1 as a late mediator of
endotoxin lethality in mice. Science 285, 248-251.

RI
Wang H., Qu, H.andDeng, H., 2015, Plasma hmgb-1 levels in subjects with obesity and type 2 diabetes:
A cross-sectional study in china. PloS one 10, e0136564.

SC
Wang L., Chen, Y., Li, X., Zhang, Y., Gulbins, E.andZhang, Y., 2016, Enhancement of endothelial
permeability by free fatty acid through lysosomal cathepsin b-mediated nlrp3 inflammasome
activation. Oncotarget 7, 73229-73241.
Weisberg S.P., McCann, D., Desai, M., Rosenbaum, M., Leibel, R.L.andFerrante, A.W., Jr., 2003,

U
Obesity is associated with macrophage accumulation in adipose tissue. The Journal of clinical
AN
investigation 112, 1796-1808.
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.andDosch, H.M., 2009,
Normalization of obesity-associated insulin resistance through immunotherapy. Nature
M

medicine 15, 921-929.


Wozniak S.E., Gee, L.L., Wachtel, M.S.andFrezza, E.E., 2009, Adipose tissue: The new endocrine
D

organ? A review article. Digestive diseases and sciences 54, 1847-1856.


Xu H., Barnes, G.T., Yang, Q., Tan, G., Yang, D., Chou, C.J., Sole, J., Nichols, A., Ross, J.S., Tartaglia,
TE

L.A.andChen, H., 2003, Chronic inflammation in fat plays a crucial role in the development of
obesity-related insulin resistance. The Journal of clinical investigation 112, 1821-1830.
Yan S.D., Schmidt, A.M., Anderson, G.M., Zhang, J., Brett, J., Zou, Y.S., Pinsky, D.andStern, D., 1994,
EP

Enhanced cellular oxidant stress by the interaction of advanced glycation end products with
their receptors/binding proteins. The Journal of biological chemistry 269, 9889-9897.
Yao Y., Wang, Y., Zhang, Z., He, L., Zhu, J., Zhang, M., He, X., Cheng, Z., Ao, Q., Cao, Y., Yang, P.,
C

Su, Y., Zhao, J., Zhang, S., Yu, Q., Ning, Q., Xiang, X., Xiong, W., Wang, C.Y.andXu, Y.,
2016, Chop deficiency protects mice against bleomycin-induced pulmonary fibrosis by
AC

attenuating m2 macrophage production. Molecular therapy : the journal of the American


Society of Gene Therapy 24, 915-925.
Ye J., Gao, Z., Yin, J.andHe, Q., 2007, Hypoxia is a potential risk factor for chronic inflammation and
adiponectin reduction in adipose tissue of ob/ob and dietary obese mice. American journal of
physiology. Endocrinology and metabolism 293, E1118-1128.
Youn J.H.andShin, J.S., 2006, Nucleocytoplasmic shuttling of hmgb1 is regulated by phosphorylation
that redirects it toward secretion. Journal of immunology 177, 7889-7897.
Zhang X., Wheeler, D., Tang, Y., Guo, L., Shapiro, R.A., Ribar, T.J., Means, A.R., Billiar, T.R., Angus,
D.C.andRosengart, M.R., 2008, Calcium/calmodulin-dependent protein kinase (camk) iv
mediates nucleocytoplasmic shuttling and release of hmgb1 during lipopolysaccharide

19
ACCEPTED MANUSCRIPT
stimulation of macrophages. Journal of immunology 181, 5015-5023.
Zhang Y., Proenca, R., Maffei, M., Barone, M., Leopold, L.andFriedman, J.M., 1994, Positional
cloning of the mouse obese gene and its human homologue. Nature 372, 425-432.
Ziegler U.andGroscurth, P., 2004, Morphological features of cell death. News in physiological sciences :
an international journal of physiology produced jointly by the International Union of
Physiological Sciences and the American Physiological Society 19, 124-128.

PT
RI
U SC
AN
M
D
TE
C EP
AC

20
ACCEPTED MANUSCRIPT
Figure legends

PT
RI
U SC
AN
Fig. 1. The composition of immune cells in lean versus obese adipose tissue. In the lean state, the
major immune cells in adipose tissue are eosinophils, Th2 cells, Treg cells, and M2-like
macrophages. Eosinophils and Th2 cells promote an anti-inflammatory milieu through the
M

secretion of IL-4, IL-10 and IL-13, supporting an M2-like phenotype of macrophages. In the obese
adipose tissue, adipocytes secret chemokines such as MCP-1, in response to proinflammatory
D

signals (e.g., TNF-α), to recruit circulating monocytes into adipose tissue, which then differentiate
into adipose tissue macrophages with an M1-like phenotype. Other than macrophages, neutrophils
TE

are also recruited from circulation probably through an inflammatory chemokine-dependent


manner. Obesity also promotes a shift in T cell populations with a decrease in Treg population and
an increase in CD4+ and CD8+ effector T cells. B cells release pathogenic IgG2c type antibodies
EP

and are also found in adipose tissue of obesity individuals. Adipocytes, mast cells, and T cells
contribute to inflammation by releasing specific adipokines and cytokines. M1: M1 macrophages;
C

M2: M2 macrophages; Treg: T regulatory cells; NEU: neutrophils; MC: mast cells; and EOS:
eosinophils.
AC

21
ACCEPTED MANUSCRIPT

PT
RI
U SC
AN
M

Fig. 2. An overview of HMGB1 in the induction and maintenance of chronic inflammation in


adipose tissue of obese subjects. In the state of obesity, elevated HMGB1 can either passively
D

released from necrotic adipocytes or actively secreted by living adipocytes, activated


TE

immunocytes, and adipose-derived stromal cells such as endothelial cells. HMGB1 activates DCs
in adipose tissue, which stimulate the expansion of inflammatory Th1 cells. Upon activation, the
recruited pDCs induce the production of type I interferons to promote macrophage M1 program.
EP

Macrophages, which play an essential role in obesity-induced inflammation, can sense the
increased HMGB1 and produce ROS, TNF-α as well as many other proinflammatory cytokines to
further impair the function of adipocyes.
C
AC

22
ACCEPTED MANUSCRIPT

PT
RI
U SC
AN
M
D
TE
EP

Fig. 3. A summary of HMGB1 signaling pathways in adipose tissue. During the development of
C

obesity-induced inflammation, RAGE and TLRs can be activated by the extracellular HMGB1
AC

(DAMP) as well as free nucleic acid molecules from necrotic adipocytes. HMGB1 is well known
to mediate RAGE, TLR2 and TLR4 activation. In addition, accumulated HMGB1 and DNA
owing to increased adipocytes death and adipose tissue remodeling can be sensed by TLR9. Upon
activation in this case, they mainly trigger NFκB activation to initiate the transcription of various
proinflammatory cytokines including TNF-α, IL-6 and IL-1β to promote inflammation in obesity.

23
ACCEPTED MANUSCRIPT
Highlights:

Obesity is characterized by a chronic, low-grade inflammation in local and systemic sites.


Both necrotic adipocytes and activated immune cells in adipose tissue account for the
increased HMGB1 level in obese subjects.
HMGB1 initiates and maintains chronic inflammatory state in adipose tissue through RAGE,
TLR2 and TLR4 signaling pathways.
HMGB1 could be a viable biomarker for early diagnosis of obesity and therapeutic target for

PT
prevention and intervention of obesity-induced inflammation.

RI
U SC
AN
M
D
TE
C EP
AC

S-ar putea să vă placă și