il-33/st2 axis in inflammation and immunopathology

11
IMMUNOLOGY IN SERBIA IL-33/ST2 axis in inflammation and immunopathology Marija Milovanovic Vladislav Volarevic Gordana Radosavljevic Ivan Jovanovic Nada Pejnovic Nebojsa Arsenijevic Miodrag L. Lukic Published online: 6 March 2012 Ó Springer Science+Business Media, LLC 2012 Abstract Interleukin-33 (IL-33), a member of the IL-1 family of cytokines, binds to its plasma membrane receptor, heterodimeric complex consisted of membrane-bound ST2L and IL-1R accessory protein, inducing NFkB and MAPK activation. IL-33 exists as a nuclear precursor and may act as an alarmin, when it is released after cell damage or as negative regulator of NFjB gene transcription, when acts in an intracrine manner. ST2L is expressed on several immune cells: Th2 lymphocytes, NK, NKT and mast cells and on cells of myeloid lineage: monocytes, dendritic cells and granulocytes. IL-33/ST2 axis can promote both Th1 and Th2 immune responses depending on the type of activated cell and microenvironment and cytokine network in damaged tissue. We previously described and discuss here the important role of IL-33/ST2 axis in experimental models of type 1 diabetes, experimental autoimmune encephalomyelitis, fulminant hep- atitis and breast cancer. We found that ST2 deletion enhance the development of T cell-mediated autoimmune disorders, EAE and diabetes mellitus type I. Disease development was accompanied by dominantly Th1/Th17 immune response but also higher IL-33 production, which suggest that IL-33 in receptor independent manner could promote the development of inflammatory autoreactive T cells. IL-33/ST2 axis has protective role in Con A hepatitis. ST2-deficient mice had more severe hepatitis with higher influx of inflammatory cells in liver and dominant Th1/Th17 systemic response. Pretreatment of mice with IL-33 prevented Con A-induced liver damage through prevention of apoptosis of hepatocytes and Th2 amplification. Deletion of IL-33/ST2 axis enhances cytotoxicity of NK cells, production of IFN-c in these cells and systemic production of IFN-c, IL-17 and TNF-a, which leads to attenuated tumor growth. IL-33 treatment of tumor-bearing mice suppresses activity of NK cells, dendritic cell maturation and enhances alternative activation of macrophages. In conclusion, we observed that IL-33 has attenuated anti-inflammatory effects in T cell-mediated responses and that both IL-33 and ST2 could be further explored as potential therapeutic targets in treatment of immune-mediated diseases. Keywords IL-33/ST2 axis Á Con A hepatitis Á EAE Á MLD-STZ diabetes Á Mouse breast cancer IL-33/ST2 axis Interleukin-33 (IL-33) is a member of the IL-1 cytokine family, originally described as a nuclear protein in cerebral arteries [1] and later as NF-HEV, a nuclear factor expressed in human high endothelial venules in secondary lymphoid organs [2]. Recently, IL-33 was identified as the ligand for the orphan receptor, ST2 (IL-1RL1). ST2 mol- ecule is a member of the IL-1 receptor family [3] that exists in two forms: a transmembrane full-length form (ST2L) and a soluble, secreted form (sST2) due to differential splicing of ST2 mRNA [4]. Soluble ST2 acts as a decoy receptor for IL-33 [5]. In normal conditions, the serum concentration of soluble ST2 is below the detectable level, but elevated level of ST2 has been reported in patients with autoimmune diseases [6], asthma [7], idiopathic pulmonary fibrosis [8], myocardial infarction and heart failure [9]. M. Milovanovic Á V. Volarevic Á G. Radosavljevic Á I. Jovanovic Á N. Pejnovic Á N. Arsenijevic Á M. L. Lukic (&) Faculty of Medicine, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia e-mail: [email protected] Miodrag L. Lukic 123 Immunol Res (2012) 52:89–99 DOI 10.1007/s12026-012-8283-9

Upload: marija-milovanovic

Post on 20-Aug-2016

217 views

Category:

Documents


0 download

TRANSCRIPT

IMMUNOLOGY IN SERBIA

IL-33/ST2 axis in inflammationand immunopathology

Marija Milovanovic • Vladislav Volarevic •

Gordana Radosavljevic • Ivan Jovanovic •

Nada Pejnovic • Nebojsa Arsenijevic • Miodrag L. Lukic

Published online: 6 March 2012

� Springer Science+Business Media, LLC 2012

Abstract Interleukin-33 (IL-33), a member of the IL-1 family of cytokines, binds to its plasma membrane receptor,

heterodimeric complex consisted of membrane-bound ST2L and IL-1R accessory protein, inducing NFkB and MAPK

activation. IL-33 exists as a nuclear precursor and may act as an alarmin, when it is released after cell damage or as

negative regulator of NFjB gene transcription, when acts in an intracrine manner. ST2L is expressed on several immune

cells: Th2 lymphocytes, NK, NKT and mast cells and on cells of myeloid lineage: monocytes, dendritic cells and

granulocytes. IL-33/ST2 axis can promote both Th1 and Th2 immune responses depending on the type of activated cell and

microenvironment and cytokine network in damaged tissue. We previously described and discuss here the important role of

IL-33/ST2 axis in experimental models of type 1 diabetes, experimental autoimmune encephalomyelitis, fulminant hep-

atitis and breast cancer. We found that ST2 deletion enhance the development of T cell-mediated autoimmune disorders,

EAE and diabetes mellitus type I. Disease development was accompanied by dominantly Th1/Th17 immune response but

also higher IL-33 production, which suggest that IL-33 in receptor independent manner could promote the development of

inflammatory autoreactive T cells. IL-33/ST2 axis has protective role in Con A hepatitis. ST2-deficient mice had more

severe hepatitis with higher influx of inflammatory cells in liver and dominant Th1/Th17 systemic response. Pretreatment

of mice with IL-33 prevented Con A-induced liver damage through prevention of apoptosis of hepatocytes and Th2

amplification. Deletion of IL-33/ST2 axis enhances cytotoxicity of NK cells, production of IFN-c in these cells and

systemic production of IFN-c, IL-17 and TNF-a, which leads to attenuated tumor growth. IL-33 treatment of tumor-bearing

mice suppresses activity of NK cells, dendritic cell maturation and enhances alternative activation of macrophages. In

conclusion, we observed that IL-33 has attenuated anti-inflammatory effects in T cell-mediated responses and that both

IL-33 and ST2 could be further explored as potential therapeutic targets in treatment of immune-mediated diseases.

Keywords IL-33/ST2 axis � Con A hepatitis � EAE � MLD-STZ diabetes � Mouse breast cancer

IL-33/ST2 axis

Interleukin-33 (IL-33) is a member of the IL-1 cytokine

family, originally described as a nuclear protein in cerebral

arteries [1] and later as NF-HEV, a nuclear factor

expressed in human high endothelial venules in secondary

lymphoid organs [2]. Recently, IL-33 was identified as the

ligand for the orphan receptor, ST2 (IL-1RL1). ST2 mol-

ecule is a member of the IL-1 receptor family [3] that exists

in two forms: a transmembrane full-length form (ST2L)

and a soluble, secreted form (sST2) due to differential

splicing of ST2 mRNA [4]. Soluble ST2 acts as a decoy

receptor for IL-33 [5]. In normal conditions, the serum

concentration of soluble ST2 is below the detectable level,

but elevated level of ST2 has been reported in patients with

autoimmune diseases [6], asthma [7], idiopathic pulmonary

fibrosis [8], myocardial infarction and heart failure [9].

M. Milovanovic � V. Volarevic � G. Radosavljevic �I. Jovanovic � N. Pejnovic � N. Arsenijevic � M. L. Lukic (&)

Faculty of Medicine, Center for Molecular Medicine and Stem

Cell Research, University of Kragujevac, Svetozara Markovica

69, 34000 Kragujevac, Serbia

e-mail: [email protected]

Miodrag L. Lukic

123

Immunol Res (2012) 52:89–99

DOI 10.1007/s12026-012-8283-9

ST2L is expressed by many hematopoietic cells, NK and

NKT cells, mast cells, monocytes, dendritic cells and

granulocytes and selectively expressed by murine and

human Th2 cells but not by Th1 lymphocytes [10]. ST2

associates with IL-1R accessory protein (IL-1RAcP) to

form an IL-33 receptor (IL-33R1) [11], and IL-33 signals

via this heterodimer. Soluble form of IL-1RAcP interacts

with the sST2–IL-33 complex to increase blocking of

IL-33 signaling [12]. There is another IL-1R family mem-

ber, SIGIRR, which, accompanied by ST2L (IL-33R2),

negatively regulates IL-33 effects [13].

The binding of IL-33 to IL-33 receptor results in the

recruitment of myeloid differentiation primary response

protein 88 (MyD88), IL-1R-associated kinase 1 (IRAK1)

and IRAK4 to the receptor complex in cytoplasmic region

of ST2, which induces activation of various signaling

proteins, including nuclear factor-jB (NF-jB), inhibitor of

NF-jB-a (IjBa) and extracellular signal-regulated kinase 1

(eRK1), eRK2, p38 and c-Jun N-terminal kinase (JNK)

leading to the induction of inflammatory mediators IL-1b,

IL-3, IL-6, TNF, IL-5 and IL-13 [14–16].

At the mRNA level, IL-33 is expressed in many organs

[11] in humans and mice. However, at the protein level, IL-

33 is mainly and constitutively expressed in epithelial and

endothelial cells [17]. Immune cells, macrophages and

dendritic cells also produce IL-33 after adequate stimulation

[18]. Pathogen-associated molecular pattern (PAMP) mol-

ecules and also cytokines TNF-a, IL-1 and IFN-c stimulate

the production of IL-33 in macrophages [19, 20]. On the

other hand, some proinflammatory cytokines such as TNF-aand IL-6 are also potent inducers of soluble ST2 (sST2) [21],

which block the effects of IL-33. Analogous to other IL-1

family cytokines, it was proposed that IL-33 becomes acti-

vated by caspase activity, but pro-IL-33 does not have a

typical cleavage site seen in pro-IL-1b and IL-18. Later

findings indicated that caspase-1, caspase-3 and caspase-7

[22–24] released after apoptotic cell death inactivated IL-33

and that IL-33 is active in pro-form. IL-33 has no leader

sequence and it is not clear, at present, how it is released

from cells [25]. It was shown that biologically active pro-IL-

33 can be released by necrotic cells as ‘‘alarmin’’ [26]. Since

IL-33 is mainly expressed in lining, epithelial and endo-

thelial cells [17] and is released after cell damage, it is

proposed to have an important role in sensing damage in

various infectious and inflammatory diseases. In the absence

of proinflammatory stimuli, IL-33 is localized to the nucleus

[2, 27]. Additionally, IL-33, as a full-length protein, can act

in an intracrine manner, translocating to the nucleus, where

it binds to the chromatin and stimulates expression of IkBa,

TNF-a, and C-REL [27, 28]. Since it has been shown that IL-

33 is released from necrotic cells of structural but not

hematopoietic origin [29], intracrine action of IL-33 could

be the main way of action in immune cells.

However, it is well known that IL-33 affects the func-

tion of cells that express ST2 molecule. IL-33 polarizes

naive T cells to produce Th2-associated cytokines IL-4,

IL-5 and IL-13 [11] and functions as a chemoattractant for

Th2 cells in vitro and in vivo [30], but also induces

secretion of proinflammatory cytokines and chemokines by

mast cells [16], basophils [31] and Th1 type cytokines from

NK and NKT cells [31, 32]. Also, IL-33 amplifies polari-

zation of alternatively activated M2 macrophages [33],

induces maturation of dendritic cells [34] and may promote

Th1-type response depending on the local cytokine milieu

[35].

IL-33/ST2 axis as the first line of defense in infection

IL-33, as a full-length molecule, is mainly expressed in

epithelial and endothelial cells [17, 36] especially in high

endothelial venules [27], and it is proposed that IL-33

serves as the first line of defense against microbes that

shape adaptive immune response. Humphreys et al. [37]

were the first to show that IL-33 protects from parasitic

infection. They demonstrated increased expression of IL-

33 mRNA in the colon of resistant, but not susceptible

mice after infection with Trichiuris muris. Exogenous IL-

33 administered to susceptible mice made them resistant to

T. muris infection. IL-33 acts as the initial innate signal of

parasite invasion to the host that polarizes adaptive

immune response toward Th2-biased response, which is

host protective in this disease. It also appears that IL-33/

ST2 axis affects significantly innate as well as adaptive

immune response, as IL-33 was unable to induce parasite

expulsion in SCID mice. However, IL-33 can exacerbate

the pathology associated with chronic T. muris infection

through T cell-independent mechanisms, probably

increasing the production of IFN-c in NK cells [2]. Regu-

latory role of IL-33 in infection is also shown during

Toxoplasma gondii infection [38]. ST2 mRNA expression

was upregulated, but IL-33 mRNA expression was not

altered, in brain lesions of mice infected with T. gondii

compared with naive mice [38]. However, ST2 knockout

mice had infection with T. gondii with more severe

pathology and increased mRNA levels of iNOS, IFN-c and

TNF-a in the central nervous system [37].

IL-33/ST2 signaling affects immune response to viruses.

The serum levels of s ST2 protein were increased in

patients infected with dengue virus [39]. Infection of mice

with Theiler’s murine encephalomyelitis virus is followed

by increased expression of IL-33 in the glia, which is fol-

lowed by enhanced innate effector functions of glial cells,

which suggested that IL-33 has important role in host

defense in the CNS [38]. The role of IL-33 in antiviral

defense was also shown in respiratory syncytial virus

90 Immunology in Serbia (2012) 52:89–99

123

infection in mice. Treatment with monoclonal ST2-specific

antibody reduced lung inflammation and disease severity in

mice with Th2 type of immune response and clinical signs

of bronchiolitis [40].

IL-33/ST2 axis in fulminant hepatitis

Recently, it has been demonstrated that both IL-33 and

soluble ST2 were elevated in sera of patients with liver

failure [41]. Therefore, we decided to dissect out the role of

IL-33/ST2 axis in liver pathology by using experimental

model of acute liver injury, concanavalin A (Con A)-

induced hepatitis. Con A-induced liver injury is well-

established murine model of T cell-mediated hepatitis

[42–45]. Intravenous injection of Con A induces massive

necrosis of hepatocytes and immune cell activation that

resembles the pathology of immune-mediated fulminant

hepatitis in humans [46].

We demonstrated that IL-33/ST2 axis has protective role

in Con A hepatitis [47] showing that ST2 knockout mice

had more sever Con A-induced hepatitis than wild-type

(WT) animals. Liver damage in ST2 knockout mice was

accompanied by intrahepatic accumulation of macro-

phages, CD4? and CD8? T lymphocytes, NK and NKT

cells, while the number of CD4?Foxp3? regulatory T cells,

that have protective role in Con A-induced hepatitis [48,

49], was reduced in ST2-deficient mice. Further, ST2-/-

mice had altered systemic immune response toward Th1/

Th17 type: we found elevated levels of systemic proin-

flammatory cytokines (TNF alpha, IFN gamma and IL-17)

and attenuated serum level of IL-4 in ST2 knockout mice.

Elevated serum level of TNF-a correlated with higher

number of TNF-a producing CD4? T cells in livers of

ST2-/- mice, probably as a consequence of increased

capacity of CD4? T cells to secrete TNF in the absence of

T1/ST2 as was reported in mice treated with anti-T1/ST2

monoclonal antibodies [50]. It is well known that CD8?

effector T cells are potent producers of IFN-c [51], and that

IFN-c-producing NK and NKT cells are major effector

cells involved in Con A-induced liver injury [52, 53].

Consistent with these findings, we showed massive infil-

tration of IFN-c-producing CD8?T, NK and NKT cells in

livers of Con A-treated ST2-/- mice. Thus, we concluded

that IFN-c, absolutely necessary in the pathogenesis of Con

A hepatitis [54–56], was mainly produced by CD8?T

lymphocytes, NK and NKT cells. In Con A hepatitis, IL-17

has been reported to be both proinflammatory and without

a direct inflammation modulating role [57, 58]. In our

study, elevated serum levels of IL-17 correlated with

severe liver damage and massive infiltration of IL-17-

producing NKT cells in the liver. Recently published data

[59] confirm that IL-17 plays a pathological role in acute

liver damage. Xu et al. [59] demonstrated that a patho-

logical role of exogenous IL-23 in Con A hepatitis depends

on the production of IL-17, suggesting that in fulminate

hepatitis IL-17 plays a pathological role.

In line with these observations, we showed that pre-

treatment of WT mice with IL-33 attenuates Con

A-induced liver injury. The injection of single dose of IL-

33 significantly reduced Con A-induced liver damage in

wild-type BALB/c mice, prevented the recruitment of

mononuclear cells into the liver and increased the influx of

T regulatory cells. Further, levels of Th1 and Th17 type

cytokines in the serum of IL-33 pretreated mice were

decreased after Con A injection. Therefore, we assume that

the main mechanism by which IL-33/ST2 pathway has a

protective role in Con A-mediated liver injury is prevention

of Th1/Th17 cell-mediated hepatic immune response.

It is well known that after Con A injection, polyclonally

activated lymphocytes, particularly CD4? T lymphocytes

and NKT cells, stick to liver sinusoidal endothelium and

destroy SECs and underlying hepatocytes [46]. IL-33 is

proposed to be released as an alarmin from necrotic cells

while caspase-1, caspase-3 and caspase-7 released after

apoptotic cell death inactivate it [22–25]. Therefore, we

decided to investigate the influence of IL-33 on the

expression of pro- and anti-apoptotic genes during Con

A-induced liver injury. Pretreatment of WT BALB/c mice

with IL-33 suppressed the activation of pro-apoptotic cas-

pase-3 and mitochondrial BAX and enhanced the expres-

sion of anti-apoptotic Bcl-2 and p-ERK leading to the

attenuation of hepatitis. We and Erhardt and Tiegs [47, 60]

suggest that during acute hepatitis IL-33 is released from

damaged LSECs and hepatocytes, activates cells that

express ST2 molecule, particularly CD4? Th2 lympho-

cytes, NKT cells and activated macrophages, shifts

immune response toward Th2 type, suppresses caspase-3

activity and enhances expression of anti-apoptotic Bcl-2

and p-ERK that limits liver damage and promotes healing

of the liver tissue (illustrated in Fig. 1].

IL-33/ST2 axis in allergy and asthma

Asthma is a chronic inflammatory disease classically

characterized by increased Th2 cytokine production. IL-33

is a strong inducer of Th2 immune responses and its role in

immunopathology of asthma had been recently reviewed

elsewhere [61, 62]. Higher expression of IL-33 and sST2

was found in sera and endobronchial biopsies from asth-

matic patients [63] as well as in mouse models of asthma

induced by ovalbumin [64, 65] compared to healthy con-

trols. In the lung tissue, IL-33 and ST2 were mainly

expressed in bronchial epithelial cells [66]. Nevertheless,

the precise role of IL-33 and ST2 in mouse models of

Immunology in Serbia (2012) 52:89–99 91

123

asthma is unknown. There are the opposite results found in

the studies using ST2-deficient mice, mice treated with

anti-ST2 proteins or IL-33-deficient mice. Massive cell

influx in the lung and airway hyper-responsiveness in a

murine model of ovalbumin-induced airway inflammation

was found after intranasal administration of IL-33 [67, 68]

and in IL-33 transgenic mice [69]. IL-33 potentiates mat-

uration of dendritic cells upregulating the expression of

CD80, CD40 and OX40L, accompanied by the release of

proinflammatory cytokines, IL-6, IL-1b, TNF-a, and IL-33

also induces allergen-specific proliferation of naive T cells.

IL-33 also affects migration of dendritic cells to the lymph

nodes, where they can contribute to the priming of Th2

cells and the induction of allergic airway inflammation

[70]. In line with these findings, IL-33 knockout mice

sensitized with ovalbumin emulsified in alum showed

attenuated recruitment of inflammatory cells to the lung

and attenuated airway hyper-responsiveness [71]. Further-

more, application of blocking anti-ST2 antibodies or ST2-

Ig fusion protein inhibited eosinophilic pulmonary

inflammation and airways hyper-responsiveness [72]. In

contrast to these reports, ST2-deficient mice were not

protected in the ovalbumin-induced airway inflammation

model [73] but have attenuated inflammation in different

model of asthma. Further, an exacerbated disease was

found in wild-type or Rag-1-/- mice that had undergone

adoptive transfer of ST2-/- ovalbumin-specific Th2 cells

[74].

The reason for these differences is not clear. It could be

due to different expression of IL1RAcP in ST2-deficient

mice. IL1RAcP forms receptor complex not only with ST2

but also with IL-1 receptor (IL1R) and amplifies the signal

[75]. Thus, if there are no ST2 molecules on cell mem-

branes, IL-1 signaling could be overamplificated, inducing

inflammation. Or, if there is no soluble ST2 that blocks IL-

33, IL-33 can bind to some other receptor and induce

inflammation.

Anaphylaxis is characterized by elevated immunoglob-

ulin-E (IgE) antibodies that signal via the high affinity Fcereceptor (FceRI) to release inflammatory mediators [14].

IL-33 is markedly elevated in the serum of patients during

an anaphylactic shock and in inflamed skin tissue of

patients with atopic dermatitis. In the presence of IgE, IL-

33 activates mast cells and directly induces degranulation

following IgE sensitization. In animals that are systemi-

cally sensitized with IgE, IL-33 administration exacerbates

antigen-induced anaphylaxis and induces the degranulation

of IgE-sensitized mast cells in the skin even in the absence

of antigen [14].

IL-33/ST2 axis in T cell-mediated autoimmunity

We explored the effects of IL-33/ST2 signaling in several

autoimmune disorders mediated by T lymphocytes. Our

findings indicate that ST2 deletion and exclusion of IL-33/

Fig. 1 Proposed role of IL-33 in tissue regeneration in Con

A-induced hepatitis. Upon Con A injection, resident liver (M1)

phagocyte Con A produces proinflammatory cytokines TNF-a, IL-12,

IL-6 and IL-1b that attract CD4? and CD8? T lymphocytes, NK and

NKT cells. These immune cells either directly or through different

soluble mediators induce apoptosis (A) or necrosis (N) of hepatocytes

(H), (gray arrow). IL-33 released from necrotic hepatocytes binds to

the ST2 receptor expressed on immune cells (Th2 and M2) and

converts immune response toward Th2 type, and stimulates secretion

of matrix metalloproteinase and arginase I from alternatively

activated macrophages that promote liver regeneration (Hr) (based

on [47, 60])

92 Immunology in Serbia (2012) 52:89–99

123

ST2 axis is accompanied by enhanced susceptibility to

dominantly T cell-mediated organ-specific autoimmune

diseases.

BALB/c mice are relatively resistant to the induction of T

cell-mediated diabetes by multiple low doses of streptozo-

tocin (MLD-STZ). In BALB/c mice deletion of ST2 mole-

cule leads to enhanced susceptibility to MLD-STZ-induced

disease as evaluated by level of glycemia and glycosuria,

number of infiltrating islet cells and b cell loss [76]. Thus,

ST2-deficient mice develop insulitis and b cell loss by

apoptosis after MLD-STZ induction of disease while there

was minimal apoptosis of beta cells and no infiltrates in the

islets of wild-type, BALB/c mice. Based on these findings,

we considered that the deletion of ST2 molecule and

exclusion of IL-33/ST2 axis alters naturally prevailing Th2

response in BALB/c mice and thus allows development of

autoreactive Th1/Th17 cell-mediated disease such as dia-

betes. We found higher expression of proinflammatory

cytokines TNF-a and IFN-c in pancreatic lymph nodes of

diabetic ST2-deficient mice in the early phase of disease,

and in later phase constantly higher expression of TNF-a.

Additionally, IL-17 was detectable in the later phase of the

disease in ST2-deficient mice, but not in the draining lymph

nodes of the WT mice. Based on these findings in MLD-STZ

diabetes, we suggest that ST2 alters Th1/Th2 balance and

leads to enhanced Th1/Th17 immune response responsible

for the destruction of beta cells and diabetes.

In another model of T cell-mediated autoimmunity,

experimental autoimmune encephalomyelitis, EAE, we

have found that ST2/IL-33 axis has important role in reg-

ulating the encephalitogenic potential of T cells. Although

BALB/c mice are resistant to EAE, ST2 deletion in BALB/

c mice is accompanied by clinically and pathologically

similar expression of EAE as in susceptible C57Bl/6 mice

(unpublished results). Deletions of ST2 gene in BALB/c

mice induced development of highly pathogenic helper

cells in the induction phase of the disease and thereafter

increase influx of these cells in CNS. In spinal cords as well

as in brains of ST2-/- mice at the peak of the disease, we

found higher numbers of CD4? lymphocytes containing

IFN-c, IL-17, TNF-a and GM-CSF, but also myeloid cells

containing IL-33, while in CNS of BALB/c mice at any

time after disease induction, there was negligible number

of inflammatory cells. The importance of ST2 molecule for

EAE development was also shown by adoptive transfer of

immune ST2-/- cells that induced the disease in BALB/c

wild-type mice as in ST2-/- mice. It appears that deletion

of ST2 gene facilitates the development of highly en-

cephalitogenic T helper cells, which can be able to transfer

the disease to WT BALB/c mice. Conversely T cells from

the draining lymph nodes of MOG35-55 immunized WT

BALB/c mice did not induce clinical disease to ST2-/- and

WT recipient. These findings also suggest that ST2

expression on immune cells in CNS is not a crucial factor

that controls inflammation in CNS tissue in EAE. Among

MOG35–55 stimulated mononuclear cells that passively

transferred EAE (ST2-/- cells), we found higher frequency

of cells that contain inflammatory cytokines (IFN-c, IL-17,

TNF-a and GM-CSF) in comparison with cells isolated from

WT mice that were unable to passively induce EAE.

Development of T helper phenotype depends on the function

of APC [77], and our next goal was to determine the effects

of ST2 molecule on APC during EAE. We found that ST2

did not affect the expression of CD80, CD86 and MHCII

markers but affected relative frequencies of different sub-

populations of DC. Inflammatory dendritic cells,

CD11c?CD11b?, that migrate to lymph nodes after initial

Th1 polarizing stimulus produce abundant IL-12 and stim-

ulate IFN-c production in T lymphocytes [78], while

CD11c?CD8? cells act as regulatory cells that suppress

CD4? T lymphocytes [79]. Draining lymph nodes of ST2-/-

BALB/c mice contain higher percentage of inflammatory

type of dendritic cells and lower percentage of

CD11c?CD8? cells. Higher percentage of myeloid cells,

CD11b?, isolated from ST2-/- BALB/c mice contains

proinflammatory cytokines such as IL-1, IL-12, IL-6 and

also IL-33. Similar results were obtained after in vitro cul-

ture of dendritic cells stimulated with TLR agonist. There

were higher amounts of IL-6 and IL-23 and lower amounts

of IL-10 in cell culture supernatants of ST2-/- DC com-

pared to WT derived DC. Based on these findings, we

assume that ST2 deletion alters polarization of APC that

secrete predominantly inflammatory cytokines that leads to

the development of highly encephalitogenic T cells.

Based on our findings, it could be suggested that ST2

gene deletion leads to the production of inflammatory

innate cytokines that induce the development of domi-

nantly inflammatory Th1/Th17 response. Additionally, the

fact that strong proinflammatory type of ST2-/- antigen-

presenting cells is accompanied by higher production of

IL-33 suggests that IL-33 exhibits intracrine inflammatory

role in APC and therefore may also promote inflammation

(illustrated in Fig 2).

IL-33/ST2 axis in anti-tumor immunity

Although there are numerous findings about the role of IL-

33 in inflammation, allergy and autoimmunity, there were

no data about the role of ST2/IL-33 axis in anti-tumor

immunity and tumor growth. Recently, we showed

importance of ST2/IL-33 axis in experimental metastatic

4T1 breast cancer model in mice [80]. ST2-/- mice had

delayed appearance of palpable primary tumor as well as

slower tumor growth and reduced number and size of

metastatic colonies in lungs and livers. ST2 deletion was

Immunology in Serbia (2012) 52:89–99 93

123

accompanied by increased number of CD4? and CD8? T

lymphocytes, enhanced cytotoxic activity in vitro of

splenocytes, NK and CD8? T lymphocytes, and also

increased the numbers of IFN-c expressing NK cells. In

contrast, number of IL-10-producing NK cells was higher

in wild-type mice after tumor inoculation, while unde-

tectable in ST2-deficient mice. ST2-/- mice also have

constitutively higher percentages of activated

CD27highCD11bhigh NK cells and CD69?KLRG- NK

cells. In vivo depletion of CD8? or NK cells revealed the

key role for NK cells in enhanced anti-tumor immunity in

ST2-/- mice.

The role of IL-33/ST2 axis on NK cells function is not

fully understood. There are reports of IL-33 to directly

stimulate [32] or indirectly amplify [31] responses of iNKT

and NK cells. However, the IL-33-dependent enhancement

of IFN-c production by these cells always required the

presence of IL-12. Our results appear to be at variance with

Fig. 2 Possible mechanism of IL-33/ST2 axis impact on autoimmune

disorder development. Autoantigen challenge of ST2-/- mice in the

presence of adjuvant (or STZ-induced release of autoantigens, not

shown) induce proinflammatory polarization of antigen-presenting

cells in the draining lymph node. ST2-/- APC after stimulation with

adjuvant produce high amount of proinflammatory cytokines IL-12,

IL-1, IL-6 (see text). These APC also contain high level of IL-33 that

possibly could, by intracrine action, potentiate production of inflam-

matory cytokines. These APC present antigen to naive T cells and

induce differentiation toward Th1 type cells that express T-bet and

produce IL-17, IFN-c, GM-CSF and TNF-a. These inflammatory T

cells pass blood–tissue barrier, enter the target tissue, secrete

inflammatory cytokines and attract other immune cells that secrete

soluble factors that damage tissue. If ST2 molecule is present in APC,

autoantigen induces limited production of inflammatory cytokines and

in these cells production of IL-10 prevails. Thus, these APC polarize

naive T cells toward Th2 type that express GATA-3. Th2 cells

produce minor amount of inflammatory cytokines, do not pass blood–

tissue barrier and therefore tissue damage is not seen

94 Immunology in Serbia (2012) 52:89–99

123

these reports [31, 32]. Possible explanation of this dis-

crepancy may be related to in vivo maturation of dendritic

cells in ST2-/- mice and their effect on NK cells [81].

Dendritic cells with mature phenotype appear to be

required for the functional maturation of NK cells [81].

Mayuzumi et al. [82] have recently demonstrated that

conventional myeloid dendritic cells from IL-33 supple-

mented cultures are immature and resistant to phenotypic

and functional maturation. Thus, it could be assumed that

in vivo lack of ST2 signaling may facilitate maturation of

dendritic cells. In fact, we have obtained data indicating

that percentage and number of CD11c?CD80highCD86high

dendritic cells were significantly higher in the local lymph

nodes of ST2-/- tumor-bearing mice compared to WT

mice [80]. Thus, it appears that IL-33/ST2 signaling

facilitates primary tumor progression and metastatic dis-

semination probably affecting cytotoxic activity and cel-

lular makeup of local lymph node and spleen, indicating an

important regulatory role of IL-33/ST2 pathway in NK

physiology and anti-tumor immunity.

It also appears that ST2 deletion affects macrophage

differentiation in tumor-bearing host. Macrophages can be

categorized into two main subsets in parallel with Th1/Th2

dichotomy. M1 macrophages (classically activated) are

induced with IFN-c and characterized as IL-12- and IL-23-

producing cells that exert enhanced cytotoxic activity

against neoplastic cells [83–87]. M1 macrophages kill

tumor cells, secrete high amounts of proinflammatory

cytokines and activate anti-tumor immune response [86,

88], while M2 macrophages play a crucial role in type 2

immune response: promoting angiogenesis, remodeling

and repairing of damaged tissues and also controlling

inflammatory response by downregulation of M1-mediated

functions [86, 89–92]. Since ST2 is constitutively expres-

sed on alternatively activated macrophages [33], we won-

dered whether IL-33/ST2 axis is involved in control of

tumor development through activity of macrophages. We

showed that target disruption of ST2 is associated with

constitutive frequency of alternatively activated (CD206?)

macrophages in the spleen [80].

Thus, IL-33 could be one of the cytokines that influence

modulation of macrophages toward M2 cells, which pro-

duce IL-10 and suppress innate and adaptive anti-tumor

immune responses.

Our recent data (unpublished results) show that exoge-

nously administrated IL-33 enhances primary 4T1 tumor

growth and inhibits innate anti-tumor immunity. It appears

that IL-33 acts as an important amplifier of the develop-

ment of alternatively activated macrophages and also

markedly reduced NK cell cytotoxic activity.

Based on our findings in mouse mammary adenocarci-

noma 4T1 cancer model [80, 93], it could be suggested that

Fig. 3 Scheme of the observed effects of IL-33/ST2 axis on tumor

growth. The effects of endogenous and also exogenous IL-33 in

mammary adenocarcinoma (4T1)-bearing hosts. IL-33 activates T

cells toward Th2 phenotype and generates relatively immature

dendritic cells that do not produce IL-12p70. Immature dendritic

cells induce T-regs that contribute to an immunosuppressive

environment and facilitate metastasis. Subsequently, IL-33/ST2

signaling could upregulate OX40L on dendritic cells leading to

induction of IL-4, and more importantly immunosuppressive IL-10-

and IL-13-producing Th2-cells that promote cancer escape. IL-33/

ST2 signaling could possibly enhance the production of tumor cells

factors with immunosuppressive activity. Also, IL-33 induces IL-10

production in NK cells and decreases their cytotoxicity. In the

absence of ST2, IL-33 produced by epithelial cells and possibly tumor

cells does not lead to the activation of Th2-associated immunosup-

pressive response. Concomitantly, IL-12 produced by classically

activated M1 macrophages leads to the maturation of DC and

induction of strong Th1-polarized immune response followed by IFN-

c production, which activates tumoricidal CTLs and NK cells. These

cells with enhanced cytotoxic activity delay 4T1 tumor growth and

the development of metastases (modified from [80, 93])

Immunology in Serbia (2012) 52:89–99 95

123

the absence of ST2 molecule decreases the immunosup-

pressive Th2-type immune response mediated by IL-33

released from epithelial, endothelial or maybe tumor cells.

Then, IL-12 produced by classically activated M1 macro-

phages promote maturation of DC and consequently strong

Th1/Th17 response that activates tumoricidal NK, NKT

cells and CD8? T cytotoxic lymphocytes (illustrated in

Fig 3). In the wild-type mice, if IL-33 is overexpressed

either endogenously or exogenously, it binds to ST2-posi-

tive Th2-polarized cells and also promotes generation of

relatively immature dendritic cells that could induce T-regs

and therefore facilitate tumor progression and metastasis.

Therapeutic potential of IL-33 and soluble ST2

IL-33 is a dual-role cytokine. It can not only promote but

also reduce inflammation depending on the tissue envi-

ronment [35]. In addition, although firstly described as

Th2-type promoting cytokine, now it is known that IL-33

has pleiotropic effects, it can contribute to development of

Th1-type of immune response as well as enhanced IL-1 and

IL-18 secretion. It implies that IL-33/ST2 axis could be

considered as therapeutic target in different diseases. IL-33

can reduce inflammation depending on the tissue context,

for example, in blood vessel inflammation associated with

atherosclerosis [36]. Unexpectedly, application of IL-33 in

established Th1/Th17 mediated inflammatory conditions

such as collagen-induced arthritis exacerbated the disease

[94]. Besides, in chronic inflammation, addition of IL-33

can promote fibrosis through enhancing the production of

cytokines such as IL-13 and factors secreted by alterna-

tively activated macrophages.

Studies with IL33-/-, ST2-/- mice and application of

different anti-ST2 antibodies in the same model of disease

indicated that absence of IL-33 do not have the same

impact on disease development/attenuation as well as

absence of ST2 molecule [95]. The fundamental mecha-

nisms of the synthesis, processing, releasing and active

form of IL-33 are still poorly defined [25]. In addition, it is

unknown whether is there any other receptor that could

bind IL-33 or weather IL-33 shares ST2 as receptor with

some other cytokine. Better understanding of these pro-

cesses is essential for the future studies of IL-33 as thera-

peutic agent.

Serum level of sST2 molecule is increased in many

inflammatory conditions with anti-inflammatory effects.

Treatment of mice with mixed Th1/Th2 type of inflam-

matory bowel disease with standard anti-TNF therapy led

to attenuation of the disease that was accompanied by

higher sST2/IL-33 ratio in serum [96]. Also, adenovirus-

mediated overexpression of soluble ST2 protects from

LPS-mediated lung injury [97]. The finding that serum

sST2 increases in response to serial injection of IL-33

indicates that ST2 is induced as a negative regulator of IL-

33 [95]. Thus, it seems that modulation of serum level of

sST2 and its relative ratio with IL-33 should be further

explored as potential therapeutic target in many inflam-

matory conditions. To this, we also added the evidence that

ST2 deletion/blocking may enhance anti-tumor immunity

and stimulate NK cell activity. This may have therapeutic

implication in immune response to viruses and experi-

mental immunotherapy of malignances.

Acknowledgments This study is supported by grants ON175069,

ON175071 and ON175103 from Ministry of Education and Science,

Republic of Serbia. We thank Dr. Andrew McKenzie for providing us

ST2 knockout mice and also thank Milan Milojevic for excellent

technical assistance.

References

1. Onda H, Kasuya H, Takakura K, Hori T, Imaizumi T, Takeuchi

T, Inoue I, Takeda J. Identification of genes differentially

expressed in canine vasospastic cerebral arteries after subarach-

noid hemorrhage. J Cereb Blood Flow Metab. 1999;19(11):1279–

88.

2. Baekkevold ES, Roussigne M, Yamanaka T, Johansen FE,

Jahnsen FL, Amalric F, Brandtzaeg P, Erard M, Haraldsen G,

Girard JP. Molecular characterization of NF-HEV, a nuclear

factor preferentially expressed in human high endothelial ven-

ules. Am J Pathol. 2003;163(1):69–79.

3. Tominaga S, Jenkins NA, Gilbert DJ, Copeland NG, Tetsuka T.

Molecular cloning of the murine ST2 gene: characterization and

chromosomal mapping. Biochim Biophys Acta. 1991;1090:1–8.

4. Bergers G, Reikerstorfer A, Braselmann S, Graninger P, Bus-

slinger M. Alternative promoter usage of the Fos-responsive gene

Fit-1 generates mRNA isoforms coding for either secreted or

membrane-bound proteins related to the IL-1 receptor. EMBO J.

1994;13:1176–88.

5. Oshikawa K, Yanagisawa K, Tominaga S, Sugiyama Y.

Expression and function of the ST2 gene in a murine model of

allergic airway inflammation. Clin Exp Allergy. 2002;32(10):

1520–6.

6. Kuroiwa K, Arai T, Okazaki H, Minota S, Tominaga S. Identi-

fication of human ST2 protein in the sera of patients with auto-

immune diseases. Biochem Biophys Res Commun. 2001;284(5):

1104–8.

7. Oshikawa K, Kuroiwa K, Tago K, Iwahana H, Yanagisawa K,

Ohno S, Tominaga SI, Sugiyama Y. Elevated soluble ST2 protein

levels in sera of patients with asthma with an acute exacerbation.

Am J Respir Crit Care Med. 2001;164(2):277–81.

8. Tajima S, Oshikawa K, Tominaga S, Sugiyama Y. The increase

in serum soluble ST2 protein upon acute exacerbation of idio-

pathic pulmonary fibrosis. Chest. 2003;124(4):1206–14.

9. Weinberg EO, Shimpo M, Hurwitz S, Tominaga S, Rouleau JL,

Lee RT. Identification of serum soluble ST2 receptor as a novel

heart failure biomarker. Circulation. 2003;107(5):721–6.

10. Xu D, Chan WL, Leung BP, Huang F, Wheeler R, Piedrafita D,

Robinson JH, Liew FY. Selective expression of a stable cell

surface molecule on type 2 but not type 1 helper T cells. J Exp

Med. 1998;187(5):787–94.

11. Schmitz J, Owyang A, Oldham E, Song Y, Murphy E, McCl-

anahan TK, Zurawski G, Moshrefi M, Qin J, Li X, Gorman DM,

96 Immunology in Serbia (2012) 52:89–99

123

Bazan JF, Kastelein RA. IL-33, an interleukin-1-like cytokine

that signals via the IL-1 receptor-related protein ST2 and induces

T helper type 2-associated cytokines. Immunity. 2005;23(5):

479–90.

12. Palmer G, Lipsky BP, Smithgall MD, Meininger D, Siu S,

Talabot-Ayer D, Gabay C, Smith DE. The IL-1 receptor acces-

sory protein (AcP) is required for IL-33 signaling and soluble

AcP enhances the ability of soluble ST2 to inhibit IL-33. Cyto-

kine. 2008;42(3):358–64.

13. Bulek K, Swaidani S, Qin J, Lu Y, Gulen MF, Herjan T, Min B,

Kastelein RA, Aronica M, Kosz-Vnenchak M, Li X. The essential

role of single Ig IL-1 receptor-related molecule/Toll IL-1R8 in

regulation of Th2 immune response. J Immunol. 2009;182(5):

2601–9.

14. Pushparaj PN, Tay HK, H’ng SC, Pitman N, Xu D, McKenzie A,

Liew FY, Melendez AJ. The cytokine interleukin-33 mediates

anaphylactic shock. Proc Natl Acad Sci USA. 2009;106(24):

9773–8.

15. Allakhverdi Z, Smith DE, Comeau MR, Delespesse G. Cutting

edge: The ST2 ligand IL-33 potently activates and drives matu-

ration of human mast cells. J Immunol. 2007;179(4):2051–4.

16. Moulin D, Donze O, Talabot-Ayer D, Mezin F, Palmer G, Gabay

C. Interleukin (IL)-33 induces the release of pro-inflammatory

mediators by mast cells. Cytokine. 2007;40(3):216–25.

17. Moussion C, Ortega N, Girard JP. The IL-1-like cytokine IL-33 is

constitutively expressed in the nucleus of endothelial cells and

epithelial cells in vivo: a novel ‘alarmin’? PLoS ONE. 2008;

3(10):e3331.

18. Ohno T, Oboki K, Morita H, Kajiwara N, Arae K, Tanaka S,

Ikeda M, Iikura M, Akiyama T, Inoue J, Matsumoto K, Sudo K,

Azuma M, Okumura K, Kamradt T, Saito H, Nakae S. Paracrine

IL-33 stimulation enhances lipopolysaccharide-mediated macro-

phage activation. PLoS ONE. 2011;6(4):e18404.

19. Liu J, Buckley JM, Redmond HP, Wang JH. ST2 negatively

regulates TLR2 signaling, but is not required for bacterial lipo-

protein-induced tolerance. J Immunol. 2010;184(10):5802–8.

20. Sweet MJ, Leung BP, Kang D, Sogaard M, Schulz K, Trajkovic

V, Campbell CC, Xu D, Liew FY. A novel pathway regulating

lipopolysaccharide-induced shock by ST2/T1 via inhibition of

Toll-like receptor 4 expression. J Immunol. 2001;166:6633–9.

21. Kumar S, Tzimas MN, Griswold DE, Young PR. Expression of

ST2, an interleukin-1 receptor homologue, is induced by proin-

flammatory stimuli. Biochem Biophys Res Commun. 1997;

235(3):474–8.

22. Talabot-Ayer D, Lamacchia C, Gabay C, Palmer G. Interleukin-

33 is biologically active independently of caspase-1 cleavage. J

Biol Chem. 2009;284(29):19420–6.

23. Luthi AU, Cullen SP, McNeela EA, Duriez PJ, Afonina IS,

Sheridan C, Brumatti G, Taylor RC, Kersse K, Vandenabeele P,

Lavelle EC, Martin SJ. Suppression of interleukin-33 bioactivity

through proteolysis by apoptotic caspases. Immunity. 2009;

31(1):84–98.

24. Ali S, Nguyen DQ, Falk W, Martin MU. Caspase 3 inactivates

biologically active full length interleukin-33 as a classical cyto-

kine but does not prohibit nuclear translocation. Biochem Bio-

phys Res Commun. 2010;391(3):1512–6.

25. Zhao W, Hu Z. The enigmatic processing and secretion of

interleukin-33. Cell Mol Immunol. 2010;7(4):260–2.

26. Haraldsen G, Balogh J, Pollheimer J, Sponheim J, Kuchler AM.

Interleukin-33 - cytokine of dual function or novel alarmin?

Trends Immunol. 2009;30(5):227–33.

27. Carriere V, Roussel L, Ortega N, Lacorre DA, Americh L, Ag-

uilar L, Bouche G, Girard JP. IL-33, the IL-1-like cytokine ligand

for ST2 receptor, is a chromatin-associated nuclear factor in vivo.

Proc Natl Acad Sci USA. 2007;104(1):282–7.

28. Ali S, Mohs A, Thomas M, Klare J, Ross R, Schmitz ML, Martin

MU. The dual function cytokine IL-33 interacts with the tran-

scription factor NF-jB to dampen NF-jB-stimulated gene tran-

scription. J Immunol. 2011;187(4):1609–16.

29. Enoksson M, Lyberg K, Moller-Westerberg C, Fallon PG, Nilsson

G, Lunderius-Andersson C. Mast cells as sensors of cell injury

through IL-33 recognition. J Immunol. 2011;186(4):2523–8.

30. Komai-Koma M, Xu D, Li Y, McKenzie AN, McInnes IB, Liew

FY. IL-33 is a chemoattractant for human Th2 cells. Eur J

Immunol. 2007;37(10):2779–86.

31. Smithgall MD, Comeau MR, Yoon BR, Kaufman D, Armitage R,

Smith DE. IL-33 amplifies both Th1- and Th2-type responses

through its activity on human basophils, allergen-reactive Th2

cells, iNKT and NK cells. Int Immunol. 2008;20(8):1019–30.

32. Bourgeois E, Van LP, Samson M, Diem S, Barra A, Roga S,

Gombert JM, Schneider E, Dy M, Gourdy P, Girard JP, Herbelin

A. The pro-Th2 cytokine IL-33 directly interacts with invariant

NKT and NK cells to induce IFN-gamma production. Eur J

Immunol. 2009;39(4):1046–55.

33. Kurowska-Stolarska M, Stolarski B, Kewin P, Murphy G, Cor-

rigan CJ, Ying S, et al. IL-33 amplifies the polarization of

alternatively activated macrophages that contribute to airway

inflammation. J Immunol. 2009;183:6469–77.

34. Rank MA, Kobayashi T, Kozaki H, Bartemes KR, Squillace DL,

Kita H. IL-33-activated dendritic cells induce an atypical TH2-

type response. J Allergy Clin Immunol. 2009;123(5):1047–54.

35. Yang Q, Li G, Zhu Y, Liu L, Chen E, Turnquist H, Zhang X, Finn

OJ, Chen X, Lu B. IL-33 synergizes with TCR and IL-12 sig-

naling to promote the effector function of CD8(?) T cells. Eur J

Immunol. 2011;41(11):3351–60.

36. Miller AM, et al. IL-33 reduces the development of atheroscle-

rosis. J Exp Med. 2008;205:339–46.

37. Humphreys NE, Xu D, Hepworth MR, Liew FY, Grencis RK. IL-

33, a potent inducer of adaptive immunity to intestinal nema-todes. J Immunol. 2008;180(4):2443–9.

38. Jones LA, Roberts F, Nickdel MB, Brombacher F, McKenzie

AN, Henriquez FL, Alexander J, Roberts CW. IL-33 receptor

(T1/ST2) signalling is necessary to prevent the development of

encephalitis in mice infected with Toxoplasma gondii. Eur J

Immunol. 2010;40(2):426–36.

39. Becerra A, Warke RV, de Bosch N, Rothman AL, Bosch I.

Elevated levels of soluble ST2 protein in dengue virus infected

patients. Cytokine. 2008;41(2):114–20.

40. Hudson CA, Christophi GP, Gruber RC, Wilmore JR, Lawrence

DA, Massa PT. Induction of IL-33 expression and activity in

central nervous system glia. J Leukoc Biol. 2008;84(3):631–43.

41. Roth GA, Zimmermann M, Lubsczyk BA, Pilz J, Faybik P, Hetz

H, Hacker S, Mangold A, Bacher A, Krenn CG, Ankersmit HJ.

Up-regulation of interleukin 33 and soluble ST2 serum levels in

liver failure. J Surg Res. 2010;163(2):79–83.

42. Xiao X, Zhao P, Rodriguez-Pinto D, Qi D, Henegariu O,

Alexopoulou L, Flavell A, Wong S, Wen L. Inflammatory reg-

ulation by TLR3 in acute hepatitis. J Immunol. 2009;183:3712–9.

43. Itoh A, Isoda K, Kondoh M, Kawase M, Kobayashi M, Tamesada

M, Yagi K. Hepatoprotective effect of syringic acid and vanillic

acid on concanavalin a-induced liver injury. Biol Pharm Bull.

2009;32:1215–9.

44. Wolf AM, Wolf D, Avila MA, Moschen AR, Berasain C, Enrich

B, Rumpold H, Tilg H. Up-regulation of the anti-inflammatory

adipokine adiponectin in acute liver failure in mice. J Hepatol.

2006;44:537–43.

45. Hanson JC, Bostick MK, Campe CB, Kodali P, Lee G, Yan J,

Maher JJ. Transgenic overexpression of interleukin-8 in mouse

liver protects against galactosamine/endotoxin toxicity. J Hepa-

tol. 2006;44:359–567.

Immunology in Serbia (2012) 52:89–99 97

123

46. Tiegs G, Hentschel J, Wendel A. A T cell-dependent experi-

mental liver injury in mice inducible by concanavalin A. J Clin

Invest. 1992;90:196–203.

47. Volarevic V, Mitrovic M, Milovanovic M, Zelen I, Nikolic I,

Mitrovic S, Pejnovic N, Arsenijevic N, Lukic ML. Protective role

of IL-33/ST2 axis in Con A-induced hepatitis. J Hepatol.

2012;56(1):26–33.

48. Erhardt A, Biburger M, Papadopoulos T, Tiegs G. IL-10, regu-

latory T cells, and Kupffer cells mediate tolerance in concanav-

alin A-induced liver injury in mice. Hepatology. 2007;45(2):

475–85.

49. Wei HX, Chuang YH, Li B, Wei H, Sun R, Moritoki Y, Gershwin

ME, Lian ZX, Tian Z. CD4 ? CD25 ? Foxp3 ? regulatory T

cells protect against T cell-mediated fulminant hepatitis in a

TGF-beta-dependent manner in mice. J Immunol. 2008;

181(10):7221–9.

50. Nagata T, Mckinley L, Peschon J, Alcorn J, Aujla S, Kolls J.

Requirement of IL-17RA in Con A induced hepatitis and negative

regulation of IL-17 production in mouse T cells. J Immunol.

2008;181:7473–9.

51. Suzuki M, Maghni K, Molet S, Shimbara A, Hamid QA, Martin

JG. IFN-gamma secretion by CD8T cells inhibits allergen-

induced airway eosinophilia but not late airway responses. J

Allergy Clin Immunol. 2002;109:803–9.

52. Dong Z, Wei H, Sun R, Tian Z. The roles of innate immune cells

in liver injury and regeneration. Cell Mol Immunol. 2007;4:

241–52.

53. Takeda K, Hayakawa Y, Van Kaer L, Matsuda H, Yagita H,

Okumura K. Critical contribution of liver natural killer T cells to

a murine model of hepatitis. Proc Natl Acad Sci USA.

2000;97:5498–503.

54. Kusters S, Gantner F, Kunstle G, Tiegs G. Interferon gamma

plays a critical role in T cell dependent liver injury in mice ini-

tiated by concanavalin A. Gastroenterology. 1996;111:462–71.

55. Robinson R, Wang J, Cripps J, Milks M, English K, Pearson T,

Gorham J. End-organ damage in a mouse model of fulminant

liver inflammation requires CD4 ? T cell production of IFN-cbut is independent of Fas. J Immunol. 2009;182:3278–84.

56. Tagawa Y, Sekikawa K, Iwakura Y. Suppression of concanavalin

A-induced hepatitis in IFN-c -/- mice, but not in TNF-a -/-

mice: role for IFN-c in activating apoptosis of hepatocytes. J

Immunol. 1997;159:1418–28.

57. Zenewicz LA, Yancopoulos GD, Valenzuela DM, Murphy A,

Karow M, Flavell RA. Interleukin-22 but not interleukin-17

provides protection to hepatocytes during acute liver inflamma-

tion. Immunity. 2007;27:647–59.

58. Wondimu Z, Santodomingo-Garzon T, Le T, Swain MG. Pro-

tective role of interleukin-17 in murine NKT cell-driven acute

experimental hepatitis. Am J Pathol. 2010;177:2334–46.

59. Xu M, Morishima N, Mizoguchi I, Chiba Y, Fujita K, Kuroda M,

Iwakura Y, Cua DJ, Yasutomo K, Mizuguchi J, Yoshimoto T.

Regulation of the development of acute hepatitis by IL-23

through IL-22 and IL-17 production. Eur J Immunol. 2011;

41(10):2828–39.

60. Erhardt A, Tiegs G. IL-33—a cytokine which balances on a

knife’s edge? J Hepatol. 2012;56(1):7–10.

61. Liew FY, Pitman NI, McInnes IB. Disease-associated functions

of IL-33: the new kid in the IL-1 family. Nat Rev Immunol.

2010;10(2):103–10.

62. Oboki K, Nakae S, Matsumoto K, Saito H. IL-33 and airway

inflammation. Allergy Asthma Immunol Res. 2011;3(2):81–8.

63. Prefontaine D, Lajoie-Kadoch S, Foley S, Audusseau S, Olivenstein

R, Halayko AJ, Lemiere C, Martin JG, Hamid Q. Increased

expression of IL-33 in severe asthma: evidence of expression

by airway smooth muscle cells. J Immunol. 2009;183(8):5094–

103.

64. Kearley J, Buckland KF, Mathie SA, Lloyd CM. Resolution of

allergic inflammation and airway hyperreactivity is dependent

upon disruption of the T1/ST2-IL-33 pathway. Am J Respir Crit

Care Med. 2009;179(9):772–81.

65. Hayakawa H, Hayakawa M, Kume A, Tominaga S. Soluble ST2

blocks interleukin-33 signaling in allergic airway inflammation.

J Biol Chem. 2007;282:26369–80.

66. Prefontaine D, Nadigel J, Chouiali F, Audusseau S, Semlali A,

Chakir J, Martin JG, Hamid Q. Increased IL-33 expression by

epithelial cells in bronchial asthma. J Allergy Clin Immunol.

2010;125(3):752–4.

67. Kondo Y, Yoshimoto T, Yasuda K, Futatsugi-Yumikura S,

Morimoto M, Hayashi N, Hoshino T, Fujimoto J, Nakanishi K.

Administration of IL-33 induces airway hyperresponsiveness and

goblet cell hyperplasia in the lungs in the absence of adaptive

immune system. Int Immunol. 2008;20(6):791–800.

68. Stolarski B, Kurowska-Stolarska M, Kewin P, Xu D, Liew FY.

IL-33 exacerbates eosinophil-mediated airway inflammation. J

Immunol. 2010;185(6):3472–80.

69. Zhiguang X, Wei C, Steven R, Wei D, Wei Z, Rong M, Zhanguo

L, Lianfeng Z. Over-expression of IL-33 leads to spontaneous

pulmonary inflammation in mIL-33 transgenic mice. Immunol

Lett. 2010;131(2):159–65.

70. Besnard AG, Togbe D, Guillou N, Erard F, Quesniaux V, Ryffel

B. IL-33-activated dendritic cells are critical for allergic airway

inflammation. Eur J Immunol. 2011;41(6):1675–86.

71. Oboki K, Ohno T, Kajiwara N, Arae K, Morita H, Ishii A, Nambu

A, Abe T, Kiyonari H, Matsumoto K, Sudo K, Okumura K, Saito H,

Nakae S. IL-33 is a crucial amplifier of innate rather than acquired

immunity. Proc Natl Acad Sci USA. 2010;107(43):18581–6.

72. Coyle AJ, Lloyd C, Tian J, Nguyen T, Erikkson C, Wang L,

Ottoson P, Persson P, Delaney T, Lehar S, Lin S, Poisson L,

Meisel C, Kamradt T, Bjerke T, Levinson D, Gutierrez-Ramos

JC. Crucial role of the interleukin 1 receptor family member T1/

ST2 in T helper cell type 2-mediated lung mucosal immune

responses. J Exp Med. 1999;190(7):895–902.

73. Kurowska-Stolarska M, Kewin P, Murphy G, Russo RC, Stolar-

ski B, Garcia CC, Komai-Koma M, Pitman N, Li Y, Niedbala W,

McKenzie AN, Teixeira MM, Liew FY, Xu D. IL-33 induces

antigen-specific IL-5 ? T cells and promotes allergic-induced

airway inflammation independent of IL-4. J Immunol. 2008;

181(7):4780–90.

74. Mangan NE, Dasvarma A, McKenzie AN, Fallon PG. T1/ST2

expression on Th2 cells negatively regulates allergic pulmonary

inflammation. Eur J Immunol. 2007;37(5):1302–12.

75. Wesche H, Korherr C, Kracht M, Falk W, Resch K, Martin MU.

The interleukin-1 receptor accessory protein (IL-1RAcP) is

essential for IL-1-induced activation of interleukin-1 receptor-

associated kinase (IRAK) and stress-activated protein kinases

(SAP kinases). J Biol Chem. 1997;272(12):7727–31.

76. Zdravkovic N, Shahin A, Arsenijevic N, Lukic ML, Mensah-

Brown EP. Regulatory T cells and ST2 signaling control diabetes

induction with multiple low doses of streptozotocin. Mol

Immunol. 2009;47(1):28–36.

77. Jiang HR, Al Rasebi Z, Mensah-Brown E, et al. Galectin-3

deficiency reduces the severity of experimental autoimmune

encephalomyelitis. J Immunol. 2009;182(2):1167–73.

78. Nakano H, Lin KL, Yanagita M, Charbonneau C, Cook DN,

Kakiuchi T, Gunn MD. Blood-derived inflammatory dendritic

cells in lymph nodes stimulate acute T helper type 1 immune

responses. Nat Immunol. 2009;10(4):394–402.

79. Vinay DS, Kim CH, Choi BK, Kwon BS. Origins and functional

basis of regulatory CD11c ? CD8 ? T cells. Eur J Immunol.

2009;39(6):1552–63.

80. Jovanovic I, Radosavljevic G, Mitrovic M, Juranic VL,

McKenzie AN, Arsenijevic N, Jonjic S, Lukic ML. ST2 deletion

98 Immunology in Serbia (2012) 52:89–99

123

enhances innate and acquired immunity to murine mammary

carcinoma. Eur J Immunol. 2011;41(7):1902–12.

81. Foti M, Granucci F, Ricciardi-Castagnoli P. A central role for

tissue-resident dendritic cells in innate responses. Trends

Immunol. 2004;25(12):650–4.

82. Mayuzumi N, Matsushima H, Takashima A. IL-33 promotes DC

development in BM culture by triggering GM-CSF production.

Eur J Immunol. 2009;39(12):3331–42.

83. Mantovani A, Sozzani S, Locati M, Allavena P, Sica A.

Macrophage polarization: tumor-associated macrophages as a

paradigm for polarized M2 mononuclear phagocytes. Trends

Immunol. 2002;23:549–55.

84. Gordon S, Taylor PR. Monocyte and macrophage heterogeneity.

Nat Rev Immunol. 2005;5:953–64.

85. Mantovani A, Sica A, Locati M. Macrophage polarization comes

of age. Immunity. 2005;23:344–6.

86. Martinez FO, Helming L, Gordon S. Alternative activation of

macrophages: an immunologic functional perspective. Annu Rev

Immunol. 2009;27:451–83.

87. Verreck FA, de Boer T, Langenberg DM, Hoeve MA, Kramer M,

Vaisberg E, Kastelein R, Kolk A, de Waal-Malefyt R, Ottenhoff

TH. Human IL-23-producing type 1 macrophages promote but

IL-10-producing type 2 macrophages subvert immunity to

(myco)bacteria. Proc Natl Acad Sci USA. 2004;101:4560–5.

88. Mantovani A, Sica A, Locati M. New vistas on macrophage

differentiation and activation. Eur J Immunol. 2007;37:14–6.

89. Solinas G, Germano G, Mantovani A, Allavena P. Tumor-asso-

ciated macrophages (TAM) as major players of the cancer-related

inflammation. J Leukoc Biol. 2009;86:1065–73.

90. Stout RD, Watkins SK, Suttles J. Functional plasticity of mac-

rophages: in situ reprogramming of tumor-associated macro-

phages. J Leukoc Biol. 2009;86:1105–9.

91. Pollard JW. Trophic macrophages in development and disease.

Nat Rev Immunol. 2009;9:259–70.

92. Mantovani A, Allavena P, Sica A. Tumor-associated macro-

phages as a prototypic type II polarized phagocyte population:

role in tumor progression. Eur J Cancer. 2004;40:1660–7.

93. Jovanovic I, Pejnovic N, Radosavljevic G, Arsenijevic N, Lukic

ML. IL-33/ST2 Axis in innate and acquired immunity to tumors.

Oncoimmunology. 2012;1:229–31.

94. Palmer G, Talabot-Ayer D, Lamacchia C, Toy D, Seemayer CA,

Viatte S, Finckh A, Smith DE, Gabay C. Inhibition of interleukin-

33 signaling attenuates the severity of experimental arthritis.

Arthritis Rheum. 2009;60(3):738–49.

95. Ohto-Ozaki H, Kuroiwa K, Mato N, Matsuyama Y, Hayakawa M,

Tamemoto H, Tominaga S. Characterization of ST2 transgenic

mice with resistance to IL-33. Eur J Immunol. 2010;40(9):

2632–42.

96. Pastorelli L, Garg RR, Hoang SB, Spina L, Mattioli B, Scarpa M,

Fiocchi C, Vecchi M, Pizarro TT. Epithelial-derived IL-33 and its

receptor ST2 are dysregulated in ulcerative colitis and in exper-

imental Th1/Th2 driven enteritis. Proc Natl Acad Sci USA.

2010;107(17):8017–22.

97. Yin H, Li XY, Yuan BH, Zhang BB, Hu SL, Gu HB, Jin XB, Zhu

JY. Adenovirus-mediated overexpression of soluble ST2 provides

a protective effect on lipopolysaccharide-induced acute lung

injury in mice. Clin Exp Immunol. 2011;164(2):248–55.

Immunology in Serbia (2012) 52:89–99 99

123