Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

SHOCK, Vol. 26, No. 2, pp.

174 Y 179, 2006

HMGB1 SIGNALS THROUGH TOLL-LIKE RECEPTOR (TLR) 4 AND TLR2

Man Yu,* Haichao Wang,* Aihao Ding,† Douglas T. Golenbock,‡ Eicke Latz,‡
Christopher J. Czura,* Matthew J. Fenton,§ Kevin J. Tracey,* and Huan Yang*
From the *Laboratories of Biomedical Science, The Feinstein Institute for Medical Research, Manhasset,
NY; † Cornell University, 1300 York Avenue, New York, NY; ‡ Department of Infectious Diseases and
Immunology, University of Massachusetts Medical School, 364 Plantation Street, LRB 370M, Worcester,
MA; and §Division of Pulmonary and Critical Care, University of Maryland School of Medicine, 685 West
Baltimore Street, Baltimore, MD

Received 2 Dec 2005; first review completed 19 Dec 2005; accepted in final form 2 Mar 2006

ABSTRACT—In response to bacterial endotoxin (e.g., LPS) or endogenous proinflammatory cytokines (e.g., TNF and
IL-1"), innate immune cells release HMGB1, a late cytokine mediator of lethal endotoxemia and sepsis. The delayed
kinetics of HMGB1 release makes it an attractive therapeutic target with a wider window of opportunity for the treatment of
lethal systemic inflammation. However, the receptor(s) responsible for HMGB1-mediated production of proinflammatory
cytokines has not been well characterized. Here we demonstrate that in human whole blood, neutralizing antibodies
against Toll-like receptor 4 (TLR4, but not TLR2 or receptor for advanced glycation end product) dose-dependently
attenuate HMGB1-induced IL-8 release. Similarly, in primary human macrophages, HMGB1-induced TNF release is dose-
dependently inhibited by anti-TLR4 antibodies. In primary macrophages from knockout mice, HMGB1 activates
significantly less TNF release in cells obtained from MyD88 and TLR4 knockout mice as compared with cells from
TLR2 knockout and wild-type controls. However, in human embryonic kidney 293 cells transfected with TLR2 or TLR4,
HMGB1 effectively induces IL-8 release only from TLR2 overexpressing cells. Consistently, anti-TLR2 antibodies dose-
dependently attenuate HMGB1-induced IL-8 release in human embryonic kidney/TLR2-expressing cells and markedly
reduce HMGB1 cell surface binding on murine macrophage Y like RAW 264.7 cells. Taken together, our data suggest that
there is a differential usage of TLR2 and TLR4 in HMGB1 signaling in primary cells and in established cell lines, adding
complexity to studies of HMGB1 signaling which was not previously expected.
KEYWORDS—HMGB1, TLR2, TLR4, signal transduction, receptor

INTRODUCTION HMGB1 and its role as a late mediator in lethal systemic


inflammation make it a potential therapeutic target. However,
Vertebrate animals have evolved an innate response to
the molecular basis for HMGB1-induced cytokine signaling in
infection and/or trauma. The response is initiated upon ac-
macrophage/monocyte is poorly understood.
tivation of pro-inflammatory cytokines, including HMGB1,
Previous studies implicated receptor for advanced glycation
released either actively from macrophage/monocyte or
end product (RAGE) as an HMGB1 receptor that mediates
passively from necrotic and damaged cells (1 Y 4). The early
neurite outgrowth during brain development and migration of
macrophage/monocyte response is associated with the release
smooth muscle cells. Binding of RAGE to HMGB1 leads to
of TNF, IL-1", and other early inflammatory mediators of
activation of NF-.B and ERK1/2, p38 and SAPK/JNK kinases
shock and tissue injury. The late responses, occurring after
(13 Y 15). In addition to RAGE, Toll-like receptor (TLR) 2 and
the early pro-inflammatory response, lead to the release
TLR4 have been implicated in HMGB1 signaling (16 Y 18).
of HMGB1 into the serum which mediates systemic in-
TLRs are highly conserved proteins that activate innate
flammation, tissue damage, and lethality (1, 4). HMGB1,
immune cells in response to a variety of endogenous and
released passively by injured or necrotic cells, acts as a pro-
exogenous stimuli (19). TLR proteins transduce signals
inflammatory cytokine and functions as a major stimulus of
through a family of cytosolic Toll adapter proteins that link
necrosis-induced inflammation (2). Administration of HMGB1
to downstream signaling cascades highly similar to those
to normal animals causes inflammatory responses including
activated by IL-1 and IL-18 (20 Y 22). Recent findings
fever, weight loss and anorexia, acute lung injury, epithelial
implicated that HMGB1 can signal via both TLR2 and
barrier dysfunction, arthritis, and death (5 Y10). Anti-HMGB1
TLR4 in neutrophils and macrophage-like RAW 264.7 cells;
treatment with antibodies or other strategies to inhibit
RAGE plays only a minor role in macrophage activation by
HMGB1 rescues mice from lethal bacterial toxin Yinduced
HMGB1 (16). Others have also shown that HMGB1 acts as a
endotoxemia or sepsis and ameliorates the severity of
mediator in hepatic ischemia-reperfusion Y induced injury via
collagen-induced arthritis (5, 11, 12). The delayed release of
a process that is TLR4 dependent (17). Mice defective in
TLR4 (C3H/Hej) developed less hepatic injury as compared
Address reprint requests to Dr. Huan Yang, The Feinstein Institute for Medical with wild-type (C3H/HeOuj) controls. However, we do not
Research, Manhasset, NY 11030. E-mail: hyang@nshs.edu. know whether TLR2 or TLR4 is involved in HMGB1
Supported by grants from The Feinstein Institute for Medical Research, GCRC, M01 signaling independently or together. These results prompted
RR018535, NIH (NIGMS), and Critical Therapeutics, Inc., (Lexington, MA) (to K.J.T.).
DOI: 10.1097/01.shk.0000225404.51320.82 us to investigate the role of TLR2 and TLR4 in HMGB1
Copyright Ó 2006 by the Shock Society signaling in primary cells and established cell lines.
174
Copyright @ 2006 by the Shock Society. Unauthorized reproduction of this article is prohibited.
SHOCK AUGUST 2006 HMGB1 SIGNALS VIA TLR4 AND TLR2 175

Our findings showed that in human whole blood, HMGB1- MyD88 dominant/negative cell lines
induced IL-8 release is dose-dependently attenuated by neutral- RAW 264.7 macrophages were transiently co-transfected with an expression
plasmid encoding a murine MyD88 Y dominant-negative mutant (corresponding to
izing antibodies against TLR4 but not TLR2 or RAGE. Similarly, amino acids 146-296), or empty vector, plus a luciferase reporter plasmid under the
in primary human macrophages, HMGB1-induced TNF release control of the NF-.B Y dependent ELAM promoter.
is dose-dependently inhibited by anti-TLR4 antibodies. In CHO reporter cell lines that constitutively express human TLR2 or TLR4 were
described previously (26, 27). These reporter lines also contain a stably transfected
macrophages from knockout mice, HMGB1 stimulated signifi- ELAM-CD25 reporter gene and express human CD25 on their surface as a
cantly less TNF release in cells from MyD88 and TLR4 knockout consequence of NF-.B activation.
mice as compared with TLR2 knockout or wild-type controls. In
contrast, in human embryonic kidney (HEK) 293 cells trans- FITC labeling of HMGB1
HMGB1 protein was labeled with fluorescein isothiocyanate (FITC) using a
fected with TLRs, HMGB1 effectively induces IL-8 release from labeling kit according to the manufacturer’s instructions (Pierce).
TLR2, but not TLR4 overexpressing cells. Consistently, anti-
TLR2 antibodies dose-dependently attenuate HMGB1-induced Generation of CHO-psF-HMGB1 cell line clone and
IL-8 release in HEK/TLR2-expressing cells and markedly reduce production of HMGB1 in CHO cells
Methods for HMGB1 isolation from mammalian CHO cells were described
HMGB1 cell surface binding on murine macrophage-like RAW previously (28). Briefly, CHO cells were transfected with plasmid psF-HMGB1
264.7 cells. Our study demonstrates that HMGB1 exhibits a using the calcium phosphate method according to the manufacturer’s instructions
differential usage of TLR2 and TLR4 in different cell types. (Gibco-BRL). The best HMGB1 secreting cell line was adapted to grow in
suspension by culturing in CHO-S-SFM II media supplemented with 2 mM
glutamine, 300 2g/mL geneticin, and 1 penicillin/streptomycin. HMGB1
secretion was approximately 5 2g/mL in the medium. HMGB1 protein was
isolated from conditioned medium by affinity purification using FLAG antibody
(ANTI-FLAG 2 affinity gel) according to the manufacturer’s instructions (Sigma).
MATERIALS AND METHODS
Confocal microscopy
Materials Macrophage-like RAW 264.7 cells were plated in 8-well slide chambers (Lab-
Cell culture media (DMEM and RPMI 1640), L-glutamine, penicillin/strepto- Tek, Nalge Nunc Internationals, Naperville, IL) and used at 70% confluence. Cells
mycin, and phosphate-buffered saline (PBS) solutions were obtained from were preincubated with anti-TLR2 or TLR4 antibodies of 25 2g/mL for 30 min at
BioWhittaker, Inc. (Walkersville, MD). Opti-MEM I medium was purchased from 4-C in Opti-MEM I medium, and FITC-labeled HMGB1 of 5 2g/mL was added
Invitrogen (Carlsbad, CA). Fetal bovine serum (FBS) was obtained from Hyclone and incubated for an additional 6 h at 4-C. Cells were then washed three times with
(Logan, UT). Geneticin (G418) and !-MEM medium were from Gibco-BRL PBS (pH 7.2) and fixed for 15 min at room temperature in 10% paraformaldehyde-
(Grand Island, NY). Polymyxin B, Triton X-114, LPS (endotoxin, Escherichia coli PBS. After fixing, cells were washed three times with PBS and mounted for
0111:B4), phenylmethylsuphonyl fluoride (PMSF), macrophage-colony stimulat- fluorescent confocal microscopy analysis.
ing factor, and nonimmune rabbit IgG and 3 FLAG peptide affinity gel were
purchased from Sigma Chemicals (St. Louis, MO). Pam2CSK4 was obtained from LPS content in HMGB1
EMC Microcollections (Catalog no. L2020, Tuebingen, Germany). Isopropyl-D-
Contaminating LPS in protein preparations was removed by phase separation
thiogalactopyranoside was purchased from Pierce Fine Chemicals (Rockford, IL).
using Triton X-114 as previously described (28). Briefly, 1/20 volume of Triton
LB broth and Chinese Hamster Ovary (CHO)-S-SFM II medium were obtained
X-114 was added to HMGB1 solution. After 10 min of gentle rotation at room
from Life Technologies (Grand Island, NY). Mouse antihuman TLR2 (TL2.1, cat
temperature, the samples were centrifuged for 10 min (8,000  g) at room
no. 16-9922) and TLR4 (HTA125, cat no. 16-9917) antibodies were from
temperature, and the top layer (containing HMGB1 proteins) was carefully
eBioscience (San Diego, CA), and mouse antihuman RAGE antibodies (cat no.
aspirated and saved. The LPS content in HMGB1 preparations was measured by
MAB5328) were from Chemicon International, Inc., (Temecula, CA). Affinity-
the Chromogenic Limulus Amebocyte lysate assay according to the manufac-
purified polyclonal antibodies against mouse TLR2 (sc-10739) and TLR4
turer’s instructions (BioWhittaker, Inc.). LPS content in HMGB1 expressed in E.
antibodies (sc-10741) were from Santa Cruz Biotech, Inc., (Santa Cruz, CA).
coli. is typically 0.6 T 0.3 pg/2g (28). For stimulation experiments, polymyxin B
Functional grade Y purified antibodies against mouse TLR2 (cat no. 16-9024) and
was added to cell culture medium at 6 U/pg LPS (5).
TLR4 (cat no. 16-9924) were from eBioscience and were used in confocal
microscopy studies. Antihuman TLR2 monoclonal antibodies (sc-21759) were
used for in vitro neutralization studies in HEK 293 cells. Antibodies were dialyzed Cytokine measurements
extensively against PBS to remove sodium azide before use in cell culture. The concentrations of TNF and IL-8 were determined using commercially
obtained enzyme-linked immunosorbent assay (ELISA) kits according to the
manufacturer’s instructions (R&D System, Inc., Minneapolis, MN).
Cell culture
Murine macrophage Y like RAW 264.7 cells and CHO cells (CHO-AA8, CRL- Antibody production
1859) were obtained from American Type Culture Collection (ATCC, Rockville, Polyclonal antibodies against HMGB1 B box were raised in rabbits (Cocalico
MD). RAW 264.7 cells were cultured in RPMI 1640 medium, and CHO cells were Biologicals, Inc., Reamstown, PA) and assayed for titer by Western blotting. IgG
cultured in !-MEM media, both supplemented with 10% heat-inactivated FBS, was purified from anti-HMGB1 antiserum using Protein A agarose according to the
2 mM glutamine, and 1 penicillin/streptomycin and grown in a humidified manufacturer’s instructions (Pierce). Neutralizing activity of anti-HMGB1 anti-
incubator with 5% CO2. Cells were used at 90% confluence, and treatment was bodies was tested as previously described (11).
carried out in Opti-MEM I medium.
HEK 293 cells stably transfected with fusion proteins consisting of cyan fluorescent
Statistical analysis
protein fused to TLR2 or TLR4 at the C-terminus (or pcDNA alone) were described
previously (23). The stably transfected HEK 293 cells were cultured in DMEM Data are presented as mean T SEM, unless otherwise stated. Differences
medium supplemented with 10% FBS, 1 penicillin/streptomycin, 2 mM glutamine, between treatment groups were determined by Student’s t test. P value less than
and 500 2g/mL geneticin and used for experiments at 90% confluence. 0.05 is considered statistically significant.
Human primary monocytes were purified by density gradient centrifugation
through Ficoll from blood donated by normal individuals to the Long Island Blood RESULTS
Bank (New York Blood Center, Melville, NY) (24). Cells were allowed to
HMGB1-stimulated IL-8 release from human whole blood
differentiate into macrophages for 7 days in complete DMEM medium containing
macrophage-colony stimulating factor (1 ng/mL) and then used in the experiments. and TNF from primary macrophages are dose-dependently
MyD88, TLR2, or TLR4 knockout mice were obtained from Dr. S. Akira inhibited by anti-TLR4 antibodies
(Osaka University, Osaka, Japan). Bone marrow Y derived macrophages were
isolated and differentiated matured to macrophages in 20% L-cell medium as
To examine the role of TLRs in HMGB1 signaling, we first
previously described (25). Cells were incubated with HMGB1 (10 2g/mL) for 24 h, used whole blood obtained from healthy volunteers. Neutralizing
and TNF released in the conditioned medium was measured. antibodies against TLR2, TLR4, or RAGE were preincubated

Copyright @ 2006 by the Shock Society. Unauthorized reproduction of this article is prohibited.
176 SHOCK VOL. 26, NO. 2 YU ET AL.

FIG. 1. HMGB1-stimulated IL-8 release from human whole blood and TNF from primary macrophages are dose-dependently inhibited by anti-
TLR4 antibodies, but not by anti-TLR2 antibodies. Blood from healthy volunteers was drawn into heparinized syringes (15 U/mL blood). Whole blood
(200 2L) was preincubated with neutralizing anti-TLR2, TLR4, or RAGE antibodies or control IgG as indicated for 30 min at 37-C. HMGB1 (5 2g/mL) was then added
and incubated for an additional 4 h in the presence of polymyxin B (350 U/mL). After incubation, plasma was harvested and IL-8 release was measured. Data are
presented as mean T SEM from three donors. *P G 0.05 versus HMGB1 alone. Peritoneal macrophages from human blood (2  105 cells/well in 48-well culture
dish) were stimulated with HMGB1 (3 2g/mL) in the presence of neutralizing anti-TLR2 and TLR4 antibodies or control IgG as indicated for 16 h. TNF release was
measured in the conditioned media. Data shown are means T SEM of five to seven experiments. *P G 0.05 versus HMGB1 alone.

with whole blood (200 2L) for 30 min, and then HMGB1 (5 2g/ CHO cells also triggered IL-8 release in a dose-dependent
mL) was added for 4 h at 37-C. We observed that HMGB1- manner, suggesting that HMGB1 signals via TLR2 in HEK
mediated IL-8 release was dose-dependently inhibited by anti- 293 cells overexpressing TLR2 (Fig. 3). Positive controls
TLR4 antibodies, but not by anti-TLR2 or RAGE antibodies using LPS, a known TLR4 agonist, stimulated IL-8 release in
(Fig. 1A). Similarly, in isolated primary human macrophages, HEK/TLR4 cells but not in TLR2 or pcDNA-transfected HEK
HMGB1-induced TNF release was dose-dependently attenuated 293 cells (Fig. 3). In contrast, Pam2CSK4, a synthetic
by anti-TLR4 antibodies (Fig. 1B), suggesting that HMGB1 lipopeptide originated from mycoplasm and a specific TLR2
signals via TLR4 in human whole blood and in macrophages. agonist, significantly stimulated IL-8 release only from TLR2
overexpressing cells, indicating that the recombinant TLR
HMGB1 induces TNF release on macrophages from proteins expressed by the HEK 293 cells are capable of
wild-type and TLR2 knockout mice, not from cells obtained responding to their ligands specifically (Fig. 3). These data
from MyD88 or TLR4 knockout mice also showed discrepancy using TLRs in HMGB1 signaling
Previous studies indicated that TLR proteins transduce between primary cells and established cell lines.
signals through an intermediate protein MyD88 (19, 29). To
HMGB1-stimulated IL-8 release is dose-dependently
determine the role of MyD88 and TLRs in HMGB1 signaling,
inhibited by anti-TLR2 antibodies in HEK/TLR2
we next used bone marrow cells obtained from wild-type,
overexpressing cells
MyD88, TLR2, or TLR4 knockout mice (25). Cells were
matured to macrophages in 20% L-cell medium and were then We used anti-TLR2 antibodies to ascertain the specificity
of TLR2-dependent HMGB1 signaling as observed in HEK/
incubated with HMGB1 (10 2g/mL) for 24 h, and TNF release
was measured (Fig. 2). HMGB1 stimulated TNF release from
wild type and TLR2 knockouts, but significantly less TNF
release occurred in the MyD88 and TLR4 knockout cells.
These data confirmed the results from human whole blood and
indicated that MyD88 and TLR4 are important in HMGB1
signaling in primary cells.

HMGB1 stimulates IL-8 release from HEK 293 cells


overexpressing TLR2, but not from HEK 293 cells
overexpressing TLR4 or vector protein
To further examine the role of TLRs in HMGB1 signaling,
we used the cultures of HEK 293 cells engineered to
overexpress either TLR2, TLR4, or control vector alone FIG. 2. HMGB1 stimulates TNF release from macrophages obtained
(23). Unexpectedly, HMGB1 (produced in E. coli) dose- from wild-type, but not MyD88, TLR2, or TLR4 knockout mice. Bone
marrow cells were obtained from wild-type, MyD88, TLR2, or TLR4 knockout
dependently stimulated IL-8 release in HEK 293 cells over- mice and were matured to macrophages in 20% L-cell medium. Cells were
expressing TLR2 but not from cells overexpressing TLR4 or incubated with HMGB1 (10 2g/mL) for 24 h, and TNF released in the
control vector alone (Fig. 3). To eliminate the possibility of conditioned medium was measured. Data are presented as percent of TNF
stimulated by wild-type cells with n = 3 separate experiments. (The range of
bacterial contamination from E. coli, we used HMGB1 HMGB1-stimulated TNF release varies from 250 to 450 pg/mL.) *P G 0.05
expressed in mammalian CHO cells. HMGB1 purified from versus wild-type control.

Copyright @ 2006 by the Shock Society. Unauthorized reproduction of this article is prohibited.
SHOCK AUGUST 2006 HMGB1 SIGNALS VIA TLR4 AND TLR2 177

FIG. 3. HMGB1 (expressed in E. coli or CHO cells)


induces IL-8 release from HEK 293 cells overexpress-
ing TLR2, but not from cells overexpressing TLR4 or
vector. HEK 293 cells overexpressing TLR2, TLR4, or
vector alone (pcDNA3) were plated in 96-well culture
plates and were used at 90% confluence. Cells were
stimulated with recombinant HMGB1 expressed in either
E. coli or mammalian CHO cells, positive controls using
LPS (ligand for TLR4, 100 ng/mL) or Pam2CSK4 (ligand
for TLR2, 1 ng/mL) for 18 h; the conditioned media were
assayed for IL-8 release by ELISA. Data represent mean T
SEM; n = 3-6 independent experiments. *P G 0.05 versus
unstimulated control.

TLR2 overexpressing cells. Neutralizing anti-TLR2 anti- DISCUSSION


bodies dose-dependently inhibited HMGB1-induced IL-8 HMGB1 released by monocyte/macrophage after exposure to
release. This suppression was specific because nonimmune TNF, IL-1", or bacterial toxins causes delayed lethality and
control IgG did not significantly inhibit HMGB1-induced IL-8 inflammation (5, 24). HMGB1 can interact with TLR2, TLR4,
release (Fig. 4). and RAGE in established cell lines and in animal models (8,
16Y18, 30). TLRs recognize specific molecular patterns that
HMGB1 interacts with TLR2 in RAW 264.7 cells are present in microbial products (19, 29). TLR4 recognizes the
To further study the interaction between TLRs and Gram-negative product LPS and participates in the recognition
HMGB1, we examined the effects of anti-TLR2 and anti- of several endogenous ligands including heat shock proteins,
TLR4 antibodies on HMGB1 cell surface binding in macro- whereas TLR2 recognizes various fungal, Gram-positive, and
phage cultures using confocal microscopy. Cell surface mycobacterial components (19, 29, 31, 32). The purpose of this
binding of FITC-HMGB1 was observed in macrophage-like study was to further examine what receptors are involved in
RAW 264.7 cells incubated with FITC-HMGB1 for 6 h at 4-C HMGB1-induced activation of inflammatory signaling using
(Fig. 5). Pretreatment with anti-TLR4 antibodies or IgG did both primary and established cell lines. We observed unex-
not have any significant effect, whereas anti-TLR2 antibodies pected difference in requirement for TLRs in HMGB1 signal-
markedly reduced HMGB1 cell surface binding as revealed by ing in different cell types: (a) HMGB1 acts through TLR4 in
the decrease in cell-associated fluorescence compared with human whole blood and primary macrophages, and (b) in
IgG-treated controls (Fig. 5), confirming the physical inter- contrast, HMGB1 signals via TLR2 in macrophage-like RAW
action of HMGB1 with TLR2 in RAW 264.7 cells. 264.7 cells and CHO cells overexpressing TLRs, although the
involvement of another TLR4-dependent cell type cannot be
HMGB1 signaling Is dependent on MyD88 in RAW 264.7 excluded (Fig. 7). Our data immediately raise the question of
and TLR2 in CHO cells transfected with TLRs why there is differential usage of TLRs in HMGB1 signaling in
We next examined the possible role of MyD88 in different cell types?
mediating HMGB1 signaling in cell lines. RAW 264.7 cells Although previous studies showed that RAGE is involved
were transfected with plasmids encoding a dominant negative in HMGB1 signaling (8, 13, 15, 30), we found that anti-
mutant MyD88 protein and an NF-.B Y dependent luciferase RAGE antibodies did not inhibit HMGB1-induced IL-8
reporter. The dominant negative MyD88 mutant significantly
inhibited the activation of NF-.B luciferase activity in-
duced by HMGB1 indicating a MyD88-dependent response
(Fig. 6A). To address the specificity of the MyD88-dependent
response, HMGB1 was added to cultures of CHO cells
engineered to constitutively express either human TLR2 or
TLR4 (Fig. 6B). These cells also contain a NF-.B Y dependent
CD25 reporter gene as previously described (26, 27).
Compared to IL-1 which stimulated CD25 expression from
both TLR2 and TLR4 expressing cells, HMGB1 failed to
increase CD25 expression in TLR4-expressing cells but
significantly stimulated CHO cells expressing TLR2. These
data indicate that HMGB1 signals via TLR2, but not TLR4 in FIG. 4. Anti-TLR2 antibodies dose-dependently inhibit HMGB1-
induced IL-8 release in HEK/TLR2 cells. HEK 293 cells overexpressing
established cell lines. Taken together, our findings show that TLR2 were plated in 96-well culture plates. Cells were stimulated with
there is a differential usage of TLR2/TLR4 in HMGB1 HMGB1 for 18 h in the presence or absence of anti-TLR2 antibodies or
signaling, depending on the cell types. HMGB1 acts as an control IgG at the concentrations indicated. Conditioned media were
collected, and IL-8 release was measured by ELISA. *P G 0.05 versus
endogenous ligand for TLR4 on primary cells, whereas it acts HMGB1 alone. Data are presented as mean T SEM from three or more
mainly through TLR2 in established cell lines (Fig. 7). separate experiments.

Copyright @ 2006 by the Shock Society. Unauthorized reproduction of this article is prohibited.
178 SHOCK VOL. 26, NO. 2 YU ET AL.

FIG. 5. Anti-TLR2 antibodies inhibit FITC-HMGB1 cell


surface binding. Macrophage-like RAW 264.7 cells were
plated in 8-well slide chambers and were used at 70%
confluence. Cells were preincubated with anti-TLR2 and TLR4
antibodies or control IgG of 25 2g/mL for 30 min at 4-C in Opti-
MEM I medium. FITC-labeled HMGB1 (and unlabeled HMGB1)
was added at 5 2g/mL followed by an additional incubation of
6 h at 4-C. Cells were then washed three times with PBS and
were fixed in 4% paraformaldehyde-PBS solution (pH 7.2) for
20 min at room temperature. After fixing, cells were washed
with PBS three times and were mounted for viewing by
fluorescent confocal microscopy. Data are representative
from five to eight experiments. Magnification, 40.

release in human whole blood assays. However, our results do the findings by Park et al. showing that HMGB1 signals
not exclude the possibility that RAGE signaling is involved in through both TLR2 and TLR4 in human neutrophils (16) and
other cell types. Additional experiments are needed to macrophages (18) and further revealed a differential usage of
determine the relative contribution of each receptor in TLR2 and TLR4 in different cell types. It remains theoret-
HMGB1 signaling. ically possible that HMGB1 binds endotoxin and bacterial
Recent findings showed that streptococcal cell wall-induced lipoproteins and functions to synergistically increase the
joint inflammation is dependent on TLR2-signaling and responses via TLR4 and TLR2. The recent study by Tsung
MyD88, demonstrating the critical role of TLR2 and MyD88 et al. suggested that even circulating HMGB1 requires intact
in sepsis and in rheumatoid arthritis where excessive amounts TLR4 for effective signaling in vivo (17). Taken together, our
of HMGB1 are produced (7, 31). Scaffidi et al. demonstrated observations demonstrate that HMGB1 signals via distinct
that necrotic cells from wild-type mice cause inflammation, pathways through TLR4 in primary cells or TLR2 in
whereas necrotic cells from HMGB1 knockout mice do not established cell lines such as RAW 264.7 cells, CHO, or
(2). It is plausible that this putative nuclear factor is HMGB1. HEK 293 cells overexpressing TLR2 and TLR4. The reasons
These results are consistent with our current findings which are not clear, but caution should be taken when interpreting
indicate that HMGB1 is a nuclear factor that causes the HMGB1 signaling results from cell lines or overexpressing
inflammatory responses and HMGB1 acts, at least partially, systems. Understanding the nature of molecular signaling by
through TLR4 and TLR2. Our results are complementary to HMGB1 may prove therapeutically beneficial in the treatment

FIG. 6. MyD88 and TLR2 are required in HMGB1-mediated cellular responses. A, MyD88 is required for HMGB1 signaling. Subconfluent RAW 264.7
cells (overexpressing either wild-type or mutant MyD88 and a NF-.B Y dependent luciferase reporter) in 24-well plates were stimulated with HMGB1 (100 ng/mL)
for 5 h. Cells were harvested and luciferase activity was measured as described. All transfections were performed in triplicate, repeated at least three times, and a
single representative experiment is shown. Data are expressed as the ratio (fold-activation) of average luciferase values from unstimulated to stimulated cells
(subtracted for background) T SD. *P G 0.05 versus control. B, TLR2 is required for HMGB1 signaling. CHO cells overexpressing TLR2 or TLR4 were seeded into
24-well tissue culture plates and used at 90% confluence. Cells were stimulated with IL-1 (10 ng/mL) or purified HMGB1 (100 ng/mL) for 18 h. Following
stimulation, cells were stained with a phycoerythrin-labeled anti-CD25 monoclonal antibody, and surface expression of CD25 was measured by flow cytometry.
Data are expressed as the ratio (fold-activation) of the percent of CD25 positive cells in unstimulated and stimulated cell populations that were gated to exclude the
lowest 5% of cells based on mean FL1 fluorescence. Data shown are from a single experiment and are representative from three separate experiments.

Copyright @ 2006 by the Shock Society. Unauthorized reproduction of this article is prohibited.
SHOCK AUGUST 2006 HMGB1 SIGNALS VIA TLR4 AND TLR2 179
13. Hori O, Brett J, Slattery T, Cao R, Zhang J, Chen JX, Nagashima M, Lundh ER,
Vijay S, Nitecki D, et al: The receptor for advanced glycation end products
(RAGE) is a cellular binding site for amphoterin. J Biol Chem 270:
25752 Y 25761, 1995.
14. Huttunen HJ, Rauvala H: Amphoterin as an extracellular regulator of cell
motility: from discovery to disease. J Intern Med 255:351 Y 366, 2004.
15. Degryse B, Bonaldi T, Scaffidi P, Muller P, Resnati M, Sanvito F, Arrigoni G,
Bianchi ME: The high mobility group (HMG) boxes of the nuclear protein
HMG1 induce chemotaxis and cytoskeleton reorganization in rat smooth
muscle cells. J Cell Biol 152:1192 Y 1206, 2001.
16. Park JS, Svetkauskaite D, He Q, Kim J, Strassheim D, Ishizaka A, Abraham E:
Involvement of TLR2 and TLR4 in cellular activation by high mobility group
box 1 protein (HMGB1). J Biol Chem 279:7370 Y 7376, 2004.
17. Tsung A, Sahai R, Tanaka H, Nakao A, Fink MP, Lotze MT, Yang H, Li J,
Tracey KJ, Geller DA, et al: The nuclear factor HMGB1 mediates hepatic injury
after murine liver ischemia-reperfusion. J Exp Med 201:1135 Y 1143, 2005.
18. Park JS, Gamboni-Robertson F, He Q, Svetkauskaite D, Kim JY, Strassheim
FIG. 7. HMGB1 uses different TLRs in signaling for stimulation of D, Sohn JW, Yamada S, Maruyama I, Banerjee A, et al: High Mobility Group
cytokine release in primary cells and established cell lines. HMGB1 Box 1 protein (HMGB1) interacts with multiple Toll like receptors. Am J
uses different TLRs for signaling in different cell types. HMGB1 signals Physiol Cell Physiol 290:C917 Y C924, 2006.
through TLR4 in human whole blood and primary macrophages obtained 19. Aderem A, Ulevitch RJ: Toll-like receptors in the induction of the innate
from MyD88, TLR2, or TLR4 knockout mice. In contrast, HMGB1 signals via immune response. Nature 406:782 Y 787, 2000.
TLR2 in macrophage-like RAW 264.7 cells and in CHO cells overexpressing 20. Bowie A, O’Neill LA: The interleukin-1 receptor-Toll-like receptor super-
TLR2 or TLR4. family: sign generators for pro-inflammatory interleukins and microbial
products. J Leukoc Biol 67:508 Y 514, 2000.
21. O’Neill LA: Signal transduction pathways activated by the IL-1 receptor/toll-
like receptor superfamily. Curr Top Microbiol Immunol 270:47 Y 61, 2002.
of human diseases where excessive amounts of HMGB1 are 22. Martin MU, Wesche H: Summary and comparison of the signaling mecha-
produced. nisms of the Toll/interleukin-1 receptor family. Biochem Biophys Acta 1592:
265 Y 280, 2002.
23. Latz E, Visintin A, Lien E, Fitzgerald KA, Monks BG, Kurt-Jones EA,
REFERENCES Golenbock DT, Espevik T: Lipopolysaccharide rapidly traffics to and from the
1. Yang H, Wang H, Czura CJ, Tracey KJ: The cytokine activity of HMGB1. Golgi apparatus with the Toll-like receptor 4-MD-2-CD14 complex in a
J Leukoc Biol 78:1 Y 8, 2005. process that is distinct from the initiation of signal transduction. J Biol Chem
2. Scaffidi P, Misteli T, Bianchi ME: Release of chromatin protein HMGB1 by 277:47834 Y 47843, 2002.
necrotic cells triggers inflammation. Nature 418:191 Y 195, 2002. 24. Andersson U, Wang H, Palmblad K, Aveberger A-C, Bloom O, Harris HE,
3. Tracey KJ, Fong Y, Hesse DG, Manogue KR, Lee AT, Kuo GC, Lowry SF, Janson A, Kokkola R, Yang H, Tracey KJ: HMG-1 stimulates proinflamma-
Cerami A: Anti-cachectin/TNF monoclonal antibodies prevent septic shock tory cytokine synthesis in human monocytes. J Exp Med 192:565 Y 570, 2000.
during lethal bacteraemia. Nature (Lond) 330:662 Y 664, 1987. 25. Shi S, Nathan C, Schnappinger D, Drenkow J, Fuortes M, Block E, Ding A,
4. Tracey KJ. Fatal sequence, the killer within. Washington D.C.: Dana Press, Gingeras TR, Schoolnik G, Akira S, et al: MyD88 primes macrophages for
2005. full-scale activation by interferon-gamma yet mediates few responses to
5. Wang H, Bloom O, Zhang M, Vishnubhakat JM, Ombrellino M, Che J, Frazier A, Mycobacterium tuberculosis. J Exp Med 198:987 Y 997, 2003.
Yang H, Ivanova S, Borovikova L, et al: HMG-1 as a late mediator of endotoxin 26. Yoshimura AE, Lien E, Toumanen R, Dziarski D, Golenbock DT: Recognition
lethality in mice. Science 285:248 Y 251, 1999. of Gram-positive bacterial cell wall components by the innate immune system
6. Abraham E, Arcaroli J, Carmody A, Wang H, Tracey KJ: HMG-1 as a occurs via Toll-like receptor 2. J Immunol 163:1 Y 5, 1999.
mediator of acute lung injury. J Immunol 165:2950 Y 2954, 2000. 27. Delude RL, Yoshimura A, Ingalls RR, Golenbock DT: Construction of a
7. Kokkola R, Sundberg E, Ulfgren A-K, Palmblad K, Li J, Wang H, Ulloa L, lipopolysaccharide reporter cell line and its use in identifying mutants
Yang H, Yan XJ, Furie R, et al: High mobility group box chromosomal protein defective in endotoxin, but not TNF-alpha signal transduction. J Immunol
1 (HMGB1)-a novel proinflammatory mediator in synovitis. Arthritis Rheum 161:3001 Y 3009, 1998.
46:2598 Y 2603, 2002. 28. Li J, Wang H, Mason JM, Levine J, Yu M, Ulloa L, Czura CJ, Tracey KJ,
8. Sappington PL, Yang R, Yang H, Tracey KJ, Delude RL, Fink MP: HMGB1 B Yang H: Recombinant HMGB1 with cytokine-stimulating activity. J Immunol
box increases the permeability of Caco-2 enterocytic monolayers and impairs Methods 289:211 Y 223, 2004.
intestinal barrier function in mice. Gastroenterology 123:790 Y 802, 2002. 29. Takeuchi O, Akira S: Toll-like receptors; their physiological role and signal
9. Taniguchi N, Kawahara K, Yone K, Hashiguchi M, Goto M: High transduction system. Int Immunopharmacol 1:625 Y 635, 2001.
mobility group box chromosomal protein 1 plays a role in the pathogenesis of 30. Huttunen HJ, Fages C, Kuja-Panula J, Ridley AJ, Rauvala H: Receptor for
rheumatoid arthritis as a novel cytokine. Arthritis Rheum 48:971 Y 981, 2003. advanced glycation end products-binding COOH-terminal motif of amphoterin
10. Agnello D, Wang H, Yang H, Tracey KJ, Ghezzi P: HMGB1, a DNA-binding inhibits invasive migration and metastasis. Cancer Res 62:4805 Y 4811, 2002.
protein with cytokine activity, induces brain TNF and IL-6 production, and 31. Joosten LA, Koenders MI, Smeets RL, Heuvelmans-Jacobs M, Helsen MM,
mediates anorexia and taste aversion. Cytokine 18:231 Y 236, 2002. Takeda K, Akira S, Lubberts E, van de Loo FA, van den Berg WB: Toll-like
11. Yang H, Ochani M, Li J, Qiang X, Tanovic M, Harris HE, Susarla SM, Ulloa L, receptor 2 pathway drives streptococcal cell wall-induced joint inflamma-
Wang H, DiRaimon R, et al: Reversing established sepsis with antagonists of tion: critical role of myeloid differentiation factor 88. J Immunol 171:
endogenous HMGB1. Proc Natl Acad Sci U S A 101:296 Y 301, 2004. 6145 Y 6153, 2003.
12. Kokkola R, Li J, Sundberg E, Aveberger AC, Palmblad K, Yang H, Tracey KJ, 32. Vabulas RM, Braedel S, Hilf N, Singh-Jasuja H, Herter S, Ahmad-Nejad P,
Andersson U, Harris HE: Successful treatment of collagen-induced arthritis in Kirschning CJ, Da Costa C, Rammensee HG, Wagner H, et al: The endoplasmic
mice and rats by targeting extracellular high mobility group box chromosomal reticulum-resident heat shock protein Gp96 activates dendritic cells via the Toll-
protein 1 activity. Arthritis Rheum 48:2052 Y 2058, 2003. like receptor 2/4 pathway. J Biol Chem 277:15107 Y 15112, 2002.

Copyright @ 2006 by the Shock Society. Unauthorized reproduction of this article is prohibited.

You might also like