ArticlePDF AvailableLiterature Review

Receptor for advanced glycation end products (RAGE) in a dash to the rescue: Inflammatory signals gone awry in the primal response to stress

Authors:

Abstract and Figures

The multiligand receptor for advanced glycation end products (RAGE) of the Ig superfamily transduces the biological impact of discrete families of ligands, including advanced glycation end products, certain members of the S100/calgranulin family, high mobility group box-1, Mac-1 (alpha(M)beta(2), CD11b/CD18), and amyloid-beta peptide and beta-sheet fibrils. Although structurally dissimilar, at least at the monomeric level, recent evidence suggests that oligomeric forms of these RAGE ligands may be especially apt to activate the receptor and up-regulate a program of inflammatory and tissue injury-provoking genes. The challenge in probing the biology of RAGE and its impact in acute responses to stress and the potential development of chronic disease is to draw the line between mechanisms that evoke repair versus those that sustain inflammation and tissue damage. In this review, we suggest the concept that the ligands of RAGE comprise a primal program in the acute response to stress. When up-regulated in environments laden with oxidative stress, inflammation, innate aging, or high glucose, as examples, the function of these ligand families may be transformed from ones linked to rapid repair to those that drive chronic disease. Identification of the threshold beyond which ligands of RAGE mediate repair versus injury is a central component in delineating optimal strategies to target RAGE in the clinic.
Content may be subject to copyright.
Receptor for advanced glycation end products (RAGE) in a
dash to the rescue: inflammatory signals gone awry in the
primal response to stress
Kevan Herold,* Bernhard Moser,
Yali Chen,
Shan Zeng,
Shi Fang Yan,
Ravichandran Ramasamy,
Jean Emond,
Raphael Clynes,
and Ann Marie Schmidt,
†,1
*Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA; and Departments of
Surgery and
Medicine, Columbia University Medical Center, New York, New York, USA
Abstract: The multiligand receptor for advanced
glycation end products (RAGE) of the Ig superfam-
ily transduces the biological impact of discrete
families of ligands, including advanced glycation
end products, certain members of the S100/cal-
granulin family, high mobility group box-1, mem-
brane-activated complex-1, and amyloid-peptide
and -sheet fibrils. Although structurally dissimi-
lar, at least at the monomeric level, recent evi-
dence suggests that oligomeric forms of these
RAGE ligands may be especially apt to activate the
receptor and up-regulate a program of inflamma-
tory and tissue injury-provoking genes. The chal-
lenge in probing the biology of RAGE and its im-
pact in acute responses to stress and the potential
development of chronic disease are to draw the line
between mechanisms that evoke repair versus
those that sustain inflammation and tissue damage.
In this review, we suggest the concept that the
ligands of RAGE comprise a primal program in the
acute response to stress. When up-regulated in en-
vironments laden with oxidative stress, inflamma-
tion, innate aging, or high glucose, as examples,
the function of these ligand families may be trans-
formed from ones linked to rapid repair to those
that drive chronic disease. Identification of the
threshold beyond which ligands of RAGE mediate
repair versus injury is a central component in de-
lineating optimal strategies to target RAGE in the
clinic. J. Leukoc. Biol. 82: 000 000; 2007.
Key Words: inflammation immunity
THE LIGAND FAMILIES OF RECEPTOR
FOR ADVANCED GLYCATION END
PRODUCTS (RAGE)
Advanced glycation end products (AGEs)
RAGE was first described as a receptor for AGEs, the products
of nonenzymatic glycation and oxidation, which form as post-
translational modifications of proteins and lipids, primarily on
lysine and arginine groups within the backbone protein. AGEs
may transform the configuration and function of the backbone
protein; depending on the site and context, AGEs may modify
long-lived protein species of the vessel wall and tissues, lead-
ing to extensive cross-linking and virtual insolubility [1–3].
AGEs, a heterogeneous group of structures, including such
specific species as carboxy methyl lysine (CML), pentosidine,
and pyralline AGEs, may form in settings such as aging,
hyperglycemia, oxidative stress, renal failure, and inflamma-
tion. AGE modifications may impart gain-of-function proper-
ties in their substrates. For example, AGE modification of
lipoproteins enhances their atherogenic potential [4, 5]. In
chronic disease, AGEs may beget further AGE formation; AGE
interaction with RAGE increases oxidative stress [6 8]. In
turn, oxidative stress increases AGE formation; mice deficient
in NADPH oxidase fail to generate the same amount of CML
AGE, as generated by wild-type mice in an inflammatory
milieu [9, 10]. Among the heterogeneous AGEs, CML AGEs
have been shown to be specific AGE ligands of RAGE [11]
(Fig. 1). In cultured endothelial cells (EC), smooth muscle
cells (SMC), and monocytes/macrophages, CML-AGE-RAGE
interaction activated NF-B and up-regulated genes linked to
inflammation. Upon infusion into wild-type mice, CML AGE
resulted in increased expression of VCAM-1 in lung tissue, a
process dependent on RAGE, as pretreatment of the animals
with anti-RAGE IgG suppressed CML AGE-mediated up-reg-
ulation of VCAM-1 [11].
AGEs are formed in diverse organisms, from bacteria, yeast,
and Drosophila to higher-order mammals as a consequence of
the inevitable production of a key pre-AGE methylglyoxal
through the metabolism of D-glucose [12–14]. AGEs are linked
integrally to damage, but are there salutary roles for AGEs?
Potential hints to innate functions for AGEs in modifying
immune responses may be inferred from findings in in vitro
analyses. AGEs may be found on the surface of aging lympho-
cytes [15, 16]. Further, findings in dendritic cells (DC) suggest
that DC glycation may promote their development but impair
1
Correspondence: Division of Surgical Science, Department of Surgery,
Columbia University Medical Center, 630 West 168th Street, P&S 17-501,
New York, NY 10032, USA. E-mail: ams11@columbia.edu
Received December 22, 2006; revised April 14, 2007; accepted April 17,
2007.
doi: 10.1189/jlb.1206751
0741-5400/07/0082-0001 © Society for Leukocyte Biology Journal of Leukocyte Biology Volume 82, August 2007 1
Uncorrected Version. Published on May 18, 2007 as DOI:10.1189/jlb.1206751
Copyright 2007 by The Society for Leukocyte Biology.
their ability to stimulate primary T cell responses [17]. Al-
though potentially injurious in impeding the organism’s re-
sponse to antigen stress, it is conceivable that in certain
settings, T cell or DC glycation may suppress untoward im-
mune responses.
S100/calgranulins
The family of S100/calgranulins is composed of multiple mem-
bers. Although not all members of the family likely bind
RAGE, increasing evidence suggests that S100/calgranulins
beyond S100A12 and S100b bind this receptor. S100/cal-
granulins may be expressed by a wide variety of cell types,
including cells linked to rapid and sustained inflammatory
responses, such as neutrophils, monocytes/macrophages, lym-
phocytes, and DC [18]. The expression of S100/calgranulins by
EC, neurons, and transformed cells suggests that a diverse
array of responses may be elicited by release of these species.
S100/calgranulins are primarily intracellular molecules; how-
ever, upon their release into the extracellular space by auto-
crine and/or paracrine interactions, they may gain new func-
tions via their ability to bind cell surface receptors such as
RAGE [19].
In vitro analyses in cultured neurons suggested that one
measure potentially distinguishing the adaptive versus injuri-
ous impact of S100b was the “dose” to which the cells were
exposed. Whereas low (nM) concentrations of S100 mediated
survival, exposure of cultured neuronal and glioma cells re-
sulted in recruitment of proinjury pathways [20].
Thus, these concepts suggest that far from being inert,
intracellular molecules, linked solely to calcium binding and
its consequences inside the cell, S100/calgranulins possess
distinct functions and importance in the biology of the cell
through homeostasis to crisis.
High mobility group box-1 (HMGB1)
Akin to S100/calgranulins, HMGB1 usually is expressed in the
intracellular space, specifically in the nucleus, where these
molecules play roles as nonhistone, DNA-binding molecules.
Like S100/calgranulins, “activation” stimuli may trigger re-
lease of HMGB1 onto the surface of highly activated cells or
directly into the extracellular space [21, 22]. It is in that
context, we propose, that HMGB1 may be freed to interact with
RAGE.
Studies reported in the mid-1990s uncovered for the first
time that HMGB1, or amphoterin, was a signal transduction
ligand of RAGE. In the first studies, RAGE and HMGB1 were
found to colocalize in the developing cerebral cortex of embry-
onic rats; cell culture analyses suggested that the HMGB1-
RAGE interaction contributed to outgrowth of neurites in neu-
rons from prenatal rat brain [23]. It is intriguing that the
HMGB1-RAGE interaction exerted its influence in migrating
cells; specifically, HMGB1 and RAGE are expressed by trans-
formed cells, and their interaction is linked to activation of cell
migration and possibly, mechanisms linked to tumor metasta-
sis, such as activation of matrix metalloproteinases (MMPs).
Studies in vivo in murine tumors indicated that administration
of a soluble receptor decoy of RAGE, sRAGE, sharply limited
local tumor growth and particularly, metastases in vulnerable
mice [24]. In parallel, activity of MMPs and MAPK activation
was reduced greatly by RAGE blockade [24].
The studies of Tracey and colleagues [25] elucidated for the
first time the provocative possibility that HGMB1 was linked to
amplification of inflammatory mechanisms as a late mediator of
the impact of endotoxin. Treatment of RAW 264.7 murine
macrophages with LPS evoked up-regulation and release of
HMGB1 8 h after incubation. In vivo, blocking antibodies to
HMGB1 protected rodents against the impact of overwhelming
sepsis [25]. Recent studies suggest the possibility that HMGB1
may not only interact with RAGE but as well, certain Toll
receptors, such as TLR2 and TLR4 [26].
DC express HMGB1 and RAGE; release of HMGB1 by these
cells provokes clonal expansion, survival, and functional po-
larization of differentiating T cells, at least in part through
activation of MAPKs and NF-B [27]. Further experimentation
reveals that at least in vitro,HMGB1 stimulates up-regulation
of CCR7 and CXCR4 chemokine receptors in DC and their
migratory ability [22]. Although these studies were limited to
the in vitro milieu, they nevertheless suggest the possibility
that the RAGE axis plays critical roles in the adaptive immune
response. These implications from cell culture analyses require
rigorous validation in vivo.
Membrane-activated complex-1 (Mac-1)
RAGE is an endothelial adhesion receptor, which mediates
direct interaction with the 2 integrin Mac-1. RAGE-Mac-1
interaction is enhanced by incubation with the proinflammatory
RAGE ligand, S100B [28]. Recent studies indicated that
HMGB1-mediated recruitment of neutrophils was dependent
on Mac-1 but not on LFA-1. In bone marrow chimera experi-
ments, Mac-1-dependent neutrophil recruitment induced by
HMGB1 required the presence of RAGE on neutrophils but not
on EC [29]. Thus, a HMGB1-dependent pathway for inflam-
Fig. 1. CML structure. Previous studies indicated that CML AGEs are
prevalent products, which accumulate in the tissues in such settings as
inflammation, hyperglycemia, and renal failure. CML adducts are specific
ligands of RAGE. The structure of this prevalent species is depicted in the
figure.
2 Journal of Leukocyte Biology Volume 82, August 2007 http://www.jleukbio.org
matory cell recruitment and activation requires the interplay
between RAGE and Mac-1. These findings establish mecha-
nisms by which RAGE and Mac-1, via HMGB1, may be linked
to acute inflammatory responses initiated by neutrophil recruit-
ment and activation.
Amyloid-peptide (A) and -sheet fibrils
In addition to AGEs, S100/calgranulins, HMGB1, and Mac-1,
RAGE is also a signal transduction receptor for Aand
-sheet fibrils [30, 31]. An emerging concept is that although
seemingly dissimilar, common features envelop many of the
ligand families of RAGE, perhaps leading to recognition by
identical or at least closely neighboring sites within the extra-
cellular domain of RAGE, specifically, but possibly not exclu-
sively, in the V-type Ig domain of RAGE [19]. Many of ligands
of RAGE are found in monomeric and oligomeric forms. Al-
though soluble, monomeric ligands interacted with RAGE in
ARPE-19 cells, their ability to stimulate signal transduction
and modulation of gene expression via RAGE was enhanced
significantly in the oligomeric state [32].
Furthermore, the ligands of RAGE may potentiate each
other’s formation and aggregation; AGE precursor species
methylglyoxal and glyoxal may increase the aggregation and
cytotoxicity of intracellular Acarboxy-terminal fragments
[33]. In addition, it has been suggested that glycation stimu-
lates amyloid formation [34, 35]. These concepts suggest that
the ligands of RAGE may indeed supermodify each other and
that the ligands may be more similar than distinct. Studies are
gaining first insights into the structure of the receptor and
providing physical evidence for the basis of ligand-RAGE
interactions [36].
SIGNAL TRANSDUCTION—CENTRAL TO THE
BIOLOGICAL IMPACT OF LIGAND-RAGE
INTERACTION
The ligands of RAGE share common properties upon their
binding to the receptor. First, among the known ligands, such
as AGEs, S100A12 and S100b, and HMGB1, Aand -sheet
fibrils, there is cross-competition in radioligand-binding assays
[11, 19]. Second, RAGE ligands bind to the V-type Ig domain,
as elucidated by radioligand-binding assays to recombinant V
domain [11]. Although we were unable to show that ligands
bound C-type domains directly, recent studies suggested hex-
americ forms of S100A12 bound the C-type Ig domain of
RAGE [36]. Third, the binding affinities, as established in
radioligand-binding assays for ligand binding to RAGE, are
quite similar in the nM range (50 nM). Fourth, extensive
evidence indicates that each of the RAGE ligands exerts its
impact as a consequence of RAGE-mediated signal transduc-
tion. The cytoplasmic domain of RAGE is essential for RAGE-
mediated changes in gene expression and cellular properties
[11, 19]. These studies suggested that the interaction of ligands
with RAGE activates signaling pathways and thus, the platform
to alter patterns of gene expression in the cell.
RAGE is expressed in multiple, distinct cell types; thus, it
is not surprising that diverse signal transduction pathways may
be impacted by RAGE. An important but not sole means by
which RAGE exerts effects on gene expression is via activation
of NF-B [7, 37], which impacts proinflammatory/prodeath and
prosurvival pathways depending on the cell type and context.
Multiple experiments have suggested that the ligand-RAGE
interaction in cells such as EC, SMC, monocytes/macrophages,
and neurons activates NF-B. Generation of reactive oxygen
species (ROS) is a key intermediate step, at least in certain
cases, as pretreatment of EC, for example, with antioxidants,
suppresses AGE-RAGE-mediated activation of NF-B [38].
RAGE-mediated activation of NADPH oxidase may account, at
least in part, for these observations [6, 8].
Multiple members of the MAPK family have been shown to
be activated by RAGE; the ligand-RAGE interaction activates
p44/p42 (ERK) MAPK, p38 MAPK, and JNK MAPK [24, 39,
40]. In addition, other studies have illustrated that AGE-
mediated activation of ras and src kinase via RAGE in SMC is
a key step in activation of NF-B [37, 41]. The specific
signaling pathways triggered by RAGE are influenced by the
context of stimulatory signals; in the setting of arterial injury,
for example, RAGE-mediated activation of JAK/STAT path-
ways critically impacts on SMC proliferation and migration
[42]. In monocytes/macrophages, NF-B is a central target of
ligand-RAGE. Recent studies suggested that in cultured mi-
croglial cells, RAGE-mediated up-regulation of cyclooxygen-
ase-2 required recruitment of cdc42/rac and JNK MAPK signal
transduction [43].
Consistent with the concept that in distinct cell types and
forms of stress, diverse signaling may be impacted by RAGE,
it has been shown that in cultured mesangial cells, AGE-
RAGE-mediated generation of ROS triggered TGF-/Smad
signaling [44]. In other cell types, cultured, primary, sensory
neurons exposed to S100 displayed increased caspase-3 activ-
ity and nuclear DNA degradation, at least in part via activation
of PI-3K signaling [45].
Under intense investigation at this time is the precise means
by which the short, highly charged, cytoplasmic domain of
RAGE signals. Earlier reports suggested that this domain
interacted with ERK MAPK [46]; however, it is unlikely that
such findings explain or underlie the diverse signal transduc-
tion repertoire of RAGE. Further, essential to establish will be
if and how distinct ligands of RAGE may stimulate specific (or
not) signaling pathways. In addition, the complexity of this
system is enhanced by the concept that the ligands of RAGE
may interact with distinct binding molecules themselves. For
example, HMGB1 may interact with TLR2 and -4; thus, it is
possible that RAGE-distinct signaling may be characteristic of
those ligands [26]. In other settings, it was suggested that
S100b activation of myotubes was independent of RAGE,
although the specific, distinct receptors were not identified
[47]. Furthermore, it has been reported that AGEs may bind
CD36 and other scavenger receptors (SRs) [48]. Taken to-
gether, these observations suggest that specific tools, such as
RAGE antagonism and RAGE null mice, would be essential in
dissecting the specific role for RAGE in biological responses.
Based on the striking ability of RAGE ligands to activate
signal transduction and thereby, alter gene expression patterns,
it was reasonable to test these concepts in vivo. Integral to the
biology of RAGE and its ligands is their up-regulation and
Herold et al. RAGE, primal responses to stress and chronic disease 3
increased accumulation in multiple biological and disease
settings. In the sections to follow, we present evidence linking
ligand-RAGE signaling to fundamental mechanisms in the
inflammatory response.
TESTING THE ROLE OF RAGE IN
INFLAMMATION—FIRST STUDIES TESTING
PHARMACOLOGICAL ANTAGONISM OF THE
LIGAND-RAGE AXIS
Our earliest work suggested that the biological repertoire of the
AGE-RAGE interaction contributed to the pathogenesis of
diabetic complications. The discovery of S100/calgranulins
and HMGB1 as putative ligands of RAGE compelled us to
consider that RAGE played roles in inflammatory responses,
even in the absence of hyperglycemia. Using sRAGE, an
extracellular ligand-binding decoy of RAGE, prepared and
purified in a baculovirus expression system, and F(ab)
2
frag-
ments of anti-RAGE IgG or anti-S100A12 IgG, experimenta-
tion revealed that blockade of ligand-RAGE suppressed the
challenge phase of footpad edema in which infiltration of
inflammatory cells and granuloma formation developed in mice
sensitized and challenged with methylated BSA. In parallel,
nuclear extracts retrieved from RAGE-antagonized mouse
foodpads revealed strikingly diminished activation of the
proinflammatory transcription factor NF-B [19]. In mice
highly vulnerable to colitis mediated by genetic deficiency of
IL-10, chronic administration of sRAGE reduced gut inflam-
mation and activation of NF-B [19]. In parallel, gene expres-
sion was altered in sRAGE-treated mice, as animals treated
with sRAGE displayed decreased levels of TNF-in plasma
[19].
These proinflammatory effects of RAGE signaling were
probed further in a murine model of bovine Type II collagen-
induced arthritis in DBA/1 mice; significant reduction in joint
swelling and erythema was noted when mice were treated with
sRAGE. In the context of inflammatory arthritis, a possible
genetic link to RAGE was uncovered by the observation that a
genetic variant of RAGE, the G82S polymorphism, was in
linkage disequilibrium with HLA-DR4. In in vitro analyses,
transfected cells expressing G82S displayed increased binding
affinity to RAGE ligand S100A12 and enhanced generation of
cytokines and MMPs upon transfection with G82S versus the
wild-type allele in the presence of S100A12 [49]. Studies in
human subjects with rheumatoid arthritis (RA) supported this
genetic link. However, based on association studies in cell
culture, future experiments are required to probe if the pres-
ence of this variant is linked to the extent or temporal appear-
ance of joint and bone destruction in RA.
RAGE AND IMMUNE RESPONSES: STUDIES
PROBING CELLULAR CONTRIBUTIONS OF
LIGAND-RAGE AXIS
The next step in probing the mechanisms linked to ligand-
RAGE signaling in immune/inflammatory consequences was to
specifically dissect the cellular mechanisms underlying these
observations. Administration of sRAGE and studies in homozy-
gous RAGE null mice revealed that the impact of sepsis
induced by cecal ligation and puncture was attenuated signif-
icantly by antagonism or deletion of RAGE [50]. When ho-
mozygous RAGE null animals were reconstituted with endo-
thelial or hematopoietic cell expression of the receptor, the
protective impact of the global deletion mutant was reversed
[50]. One interpretation of these findings was that RAGE was
linked to amplification of inflammation in sepsis but not to the
adaptive immune response. Studies using distinct, cell-specific
mutants of RAGE, however, suggest that in specific settings,
RAGE may play roles in adaptive immune mechanisms.
RAGE and the pathogenesis of Type 1 diabetes:
studies in NOD mice
Roles for RAGE in inflammatory signaling were further studies
in NOD/scid mice subjected to adoptive transfer of diabeto-
genic spleen cells. Compared with baseline, pancreata from
diabetic NOD/scid mice, which had received a transfer of
splenocytes, revealed marked up-regulation of RAGE and
S100 on islet cells containing an inflammatory infiltrate. In
parallel, it was shown that RAGE was also expressed in a
population of T cells (CD4and CD8) and B cells [51].
To test the potential role of RAGE in mediating autoimmune
diabetes, NOD/scid mice receiving a transfer of splenocytes
from a diabetic NOD donor were treated with sRAGE or
vehicle, murine serum albumin. Animals treated with sRAGE
displayed significant reduction in the rate of transfer of diabe-
tes. By Day 36 after transfer, 92% of control animals but only
10% of mice treated with sRAGE became diabetic. In parallel,
gene expression patterns were altered when RAGE signaling
was impacted; levels of cytokines IL-1and TNF-were
reduced significantly in the sRAGE-treated islets compared
with murine serum albumin-treated animals. The expression of
IL-10 was increased in sRAGE-treated mice islets, along with
increased TGF-, compared with vehicle-treated animals [51].
In contrast to injection of diabetogenic splenocytes, when
preactivated, diabetogenic BDC2.5 cells were injected into
mice, sRAGE displayed no effect on prevention of diabetes
[51]. These investigations suggested that the ligand-RAGE axis
contributed, in part, to T lymphocyte priming or cognate DC-T
lymphocyte interactions. Studies are underway to address these
concepts; first studies in cultured T cells suggest that blockade
of RAGE attenuates T lymphocyte proliferation in response to
allostimulation [52].
Roles for RAGE in T lymphocyte responses in
the adaptive immune response
Prompted by these findings in vitro, we probed roles for RAGE
in T lymphocytes in an established model of heterotopic,
allogeneic heart transplantation, wherein fully mismatched
grafts (donor, CD1 strain, H2
q
) were transplanted into C57BL/6
recipients (H2
b
). Mice were treated with sRAGE, 100 or 200
g/day, beginning 1 day prior to transplantation and continued
once daily until sacrifice. Control animals received equal vol-
umes of PBS. The mean graft survival time in vehicle (PBS)-
treated mice was 7.3 0.7 days. Mice treated with sRAGE,
100 g/day, displayed significantly increased graft survival,
4 Journal of Leukocyte Biology Volume 82, August 2007 http://www.jleukbio.org
11.7 1.7 days. In animals treated with the higher dose of
sRAGE, 200 g/day, graft survival time was even greater, at
19.5 2.8 days [52].
Immunofluorescence microscopy revealed a significant re-
duction in cells expressing RAGE in sRAGE-treated graft
recipients. In parallel, S100 and HMGB1-expressing cell
staining was reduced significantly in hosts that received
sRAGE. Inflammation was, in turn, reduced significantly in the
sRAGE-treated grafts. Compared with PBS-treated animals,
mice treated with sRAGE displayed significantly less edema
and inflammatory cell infiltration, including reduced numbers
of T lymphocytes [52].
The finding that significantly less T cells were present in the
sRAGE-treated allografts led us to test the hypothesis that
RAGE modulated alloimmune responses directly. We used
purified T cells and MHC Class II
APC retrieved from MHC-
mismatched mice and used sRAGE and blocking antibodies to
the receptor. Incubation with sRAGE resulted in a statistically
significant, dose-dependent decrease in lymphocyte prolifera-
tion versus IgG control-treated cultures. To substantiate the
specific role of RAGE, cells in the mouse allogeneic mixed
lymphocyte culture were incubated with blocking antibodies to
RAGE. Compared with nonimmune IgG, incubation with
monoclonal anti-RAGE IgG resulted in a statistically signifi-
cant, dose-dependent decrease in lymphocyte proliferation
[52].
These findings suggested that sRAGE suppressed donor-
reactive, T cell-priming responses and led us to test the effect
of RAGE antagonism on human lymphocyte proliferation trig-
gered by alloresponses. Incubation with sRAGE resulted in a
statistically significant decrease in lymphocyte proliferation
versus control-treated cultures. Similar effects were noted with
anti-RAGE IgG. We propose that ligands released from or
presented on the surface of the irradiated cells provided the
stimulus for proliferation and activation of lymphocytes [52].
Studies are underway at this time to elucidate the precise
signaling mechanisms impacted by RAGE in T cell-prolifera-
tive and cytokine responses.
Further studies suggested the importance of RAGE in mod-
ulating T cell infiltration into immune/inflammatory sites. In a
murine model of multiple sclerosis, experimental autoimmune
encephalomyelitis (EAE), RAGE, and its inflammatory ligand
S100 were overexpressed. Blockade of RAGE, using sRAGE,
suppressed EAE disease induced by myelin basic protein
(MBP) peptide or encephalitogenic T cells or when EAE oc-
curred spontaneously in the TCR-transgenic mice devoid of
endogenous TCR-and TCR-chains. In these studies, the
striking impact of RAGE antagonism was evident by markedly
decreased infiltration of the spinal cord by immune and in-
flammatory cells. In parallel, nuclear extracts retrieved from
spleen tissue revealed marked reduction in activation of NF-
B, thereby suggesting that RAGE-mediated signaling ac-
counted, at least in part, for genes in gene expression and
cellular properties [53].
Thus, to address the role of CD4 T cell RAGE signaling
directly in these processes, transgenic mice were generated
with targeted overexpression of dominant-negative (DN) RAGE
in CD4T cells. DN RAGE consists of the extracellular and
membrane-spanning domain of RAGE; solely, the RAGE cy-
tosolic domain is deleted. Thus, although RAGE ligands may
bind the truncated receptor, they are unable to affect signal
transduction via RAGE. Compared with wild-type littermate
mice, transgenic CD4 DN RAGE mice were resistant to MBP-
induced EAE [53]. These data reinforced the significant impact
of CD4T cell signaling in RAGE-dependent, adaptive im-
mune responses and underscored roles specifically for RAGE
in mediating migration of effector cells into immune foci.
To further probe the specific mechanisms linking RAGE to
T cell responses, we recently used OT-II T cells reactive with
OVA. Preliminary studies using RAGE-expressing versus
RAGE null OT-II cells suggest that RAGE is required for
effective T cell priming. Experiments are underway to delin-
eate the specific signal transduction mechanisms underlying
these findings [54].
Roles for RAGE in DC/macrophage responses in
the adaptive immune response
Extensive studies, particularly in vitro, highlighted roles for
RAGE signaling in monocyte/macrophage migration and acti-
vation, the latter as defined by up-regulation of proinflamma-
tory factors such as cytokines and MMPs. These concepts were
probed in vivo in a murine model of massive liver injury. The
ability of the liver to regenerate is finite; in experimental
systems, 70% resection of the liver triggers a fully effective
regeneration program in which liver mass is restored in parallel
with function. However, in contrast, when 20% more liver
tissue is removed (85% resection), the threshold for recruit-
ment of effective regeneration programs is exceeded in rodents,
such that overwhelming inflammation and apoptosis of the
remnant ensue. Given the strong link to inflammatory mecha-
nisms in this setting, we probed the role of the RAGE axis.
The first suggestion that RAGE might be implicated in
massive liver injury was the observation that consequent to
resections, RAGE mRNA transcripts were up-regulated selec-
tively in the 85% but not 70% resection setting. Immunohis-
tochemistry using anti-RAGE IgG revealed the intriguing find-
ing that the principal site of RAGE expression in the remnant
after massive resection was in cells expressing CD11c and
CD68, thus suggesting that RAGE was expressed largely in
mononuclear phagocyte (MP)-derived DC (MPDDC) [55].
These observations led us to posit that MPDDC, in part via
RAGE, modulated inflammatory responses in the liver rem-
nant, which contributed to massive apoptosis and failure of
regeneration.
To address these concepts, we first administered sRAGE to
the animals undergoing massive resection. Compared with
vehicle-treated mice, a significant increase in survival of wild-
type C57BL/6 mice was noted in sRAGE-treated mice; the
effects of sRAGE were dose-dependent. To target RAGE and
its inflammatory ligands directly, we prepared F(ab)
2
frag-
ments of anti-RAGE IgG, anti-S100 IgG, and anti-HMGB1
IgG. Compared with administration of nonimmune F(ab)
2
frag-
ments, mice treated with anti-RAGE, anti-S100, or anti-
HMGB1 F(ab)
2
fragments displayed significantly increased
survival. Consistent with the concept that sRAGE trapped
RAGE ligands, we found that when plasma of sRAGE-treated
mice was subjected to immunoprecipitation with anti-RAGE
IgG, followed by immunoblotting of bound material with anti-
Herold et al. RAGE, primal responses to stress and chronic disease 5
bodies to S100/calgranulin, S100/calgranulin epitopes were
revealed. Thus, studies using sRAGE and these antiligand
antibody fragments strongly suggested that the ligand-RAGE
axis contributed to impaired survival and failure of regenera-
tion in these remnants [55].
In liver resection, it is well-established that inflammation
must be tempered appropriately, such that proregenerative and
not prodeath pathways would be recruited selectively. Our
findings revealed that administration of sRAGE modulated
cytokine expression in the remnant and drove early up-regu-
lation of NF-B activity, in parallel, with reduction in TUNEL-
expressing cells and decreased activation of caspase-3. As
sRAGE and anti-RAGE F(ab)
2
fragments would be expected
to target the remnant globally, we prepared transgenic mice in
which RAGE signaling would be mutated in cells of MP
lineage, including cells expressing CD11c and CD68, as driven
by the macrophage SR Type A (SR-A) promoter, referred to as
SR DN RAGE mice. Compared with wild-type mice, we found
that transgenic SR DN RAGE mice displayed significantly
improved survival, regeneration of the hepatic remnant, mod-
ulation of proregenerative cytokines, and early up-regulation of
NF-B activity. It is important to note that by using this
promoter, it was not possible to dissect the specific impact of
infiltrating monocytes/macrophages, Kupffer cells, or MPDDC.
However, it was evident that blunting RAGE impact in cells of
MP lineage restored effective regeneration, even in the face of
massive resection of the liver [55].
Thus, in these studies, blockade of RAGE in massive liver
resection restored beneficial, inflammatory responses and ac-
tivation of NF-B, suggesting that RAGE played key roles in
modulation of these central pathways in inflammatory mecha-
nisms. These considerations prompted us to probe the hypoth-
esis that at least in certain settings, RAGE might contribute to
beneficial, inflammatory mechanisms.
RAGE and inflammatory mechanisms linked to
nerve regeneration
The specific hypothesis that RAGE-dependent mechanisms
might mediate adaptive repair as a consequence of inflamma-
tory signaling was addressed in a murine model of unilateral
crush of the sciatic nerve. Upon crush of the nerve, rapid
recruitment of proinflammatory mechanisms ensues, which
contributes to adaptive remodeling in the crushed nerve seg-
ments, along with up-regulation of regenerative pathways. Our
studies revealed that RAGE and its ligands, particularly S100/
calgranulins and HMGB1, were up-regulated rapidly within
hours of sciatic nerve crush. RAGE expression was up-regu-
lated in macrophages and in axonal elements within the
crushed nerve segment [56]. As these experiments placed
RAGE and its ligands at the site of peripheral nerve crush, it
was logical to explore the outcome of RAGE blockade in this
context. Did RAGE contribute to repair versus failure of re-
generation?
Fig. 2. RAGE and key roles in acute stress versus pathways linked to chronic disease— hypotheses and unifying concepts. We hypothesize that the ligands of
RAGE possess adaptive roles in primal responses to short and self-limiting stresses, which accompany host existence within their environment. In uncomplicated
environments, such stresses may promote release of RAGE ligands, largely in monomeric forms, in a manner linked to their rapid engagement of primarily innate
immune receptors and to a degree, RAGE. Rapid detoxification and removal of these ligands after the burst of release and response to stress ensure repair and
return to homeostasis. In contrast, in complex settings, chronic inflammation, hyperglycemia, and innate aging prime these ligands to undergo supermodification,
in which oligomeric forms may predominate. We predict that under such conditions, these ligand configurations recognize and activate RAGE preferentially and
chronically. In turn, the consequences of such interactions favor long-term tissue stress. Identification of strategies to retain primal RAGE responses to stress, yet
derail amplification pathways, which damage tissues irrevocably, holds great promise in harnessing lessons learned from the biology of RAGE to clinical trials.
6 Journal of Leukocyte Biology Volume 82, August 2007 http://www.jleukbio.org
To address these questions, we administered sRAGE or
blocking F(ab)
2
fragments derived from anti-RAGE IgG to
wild-type mice subjected to acute crush to the sciatic nerve.
Motor and sensory conduction velocities, walking tract analy-
ses, and regeneration, as assessed by myelinated fiber densi-
ties, were impaired in RAGE-blocked mice. Histological and
molecular analysis revealed that macrophage infiltration and
thus, Wallerian degeneration were reduced in RAGE-blocked
mice [56]. To determine if macrophage and/or neuronal RAGE
signaling participated critically in the response to nerve crush,
transgenic mice expressing DN RAGE in macrophages or
peripheral neurons, driven by the SR-A or thy-1 promoter,
respectively, were used. Compared with littermate controls,
regeneration was delayed in the transgenic mouse but espe-
cially, in double-transgenic mice expressing DN RAGE in
macrophages and peripheral neurons [57].
These studies were the first to demonstrate that recruitment
of RAGE facilitated beneficial inflammatory mechanisms. Cer-
tainly, however, the biology of tissue repair in the context of
RAGE is complex, as administration of sRAGE to diabetic
mice subjected to full-thickness excisional wounds accelerated
wound healing, largely by suppression of exaggerated inflam-
matory mechanisms and blunting of excessive MMP activity. In
contrast, when sRAGE was administered to nondiabetic mice,
no impact, beneficial or deleterious, was observed on wound
closure, perhaps as the wounds close quite rapidly [58]. Fur-
ther, in bacterially challenged mice, periodontal wound healing
was improved in diabetic mice treated with sRAGE; in parallel,
cytokine generation and MMP activity in gingival tissue were
reduced significantly [59].
PERSPECTIVES AND HYPOTHESES
In the full context of the biological response to stress, exquisite
control of inflammatory mechanisms is critical to executing the
best balance between rapid resolution of injury versus ampli-
fication of proinjury pathways. Thus, it is not surprising that
multiple receptors and pathways and their cross-talk must be
involved in the primal response to acute stress (Fig. 2). In the
biology of RAGE, we propose that the ligand families, which
may mediate rapid repair in such responses, are likely one and
the same, sustaining and mediating chronic tissue perturbation
and injury. We predict that the distinction will lie in the
specific environments and forms of presentation of RAGE
ligands. Common to many of the ligand families of RAGE is the
ability to form oligomers, rendering them more apt to bind and
activate RAGE. Environmental modulation of the ligand fam-
ilies, such as in chronic diseases beset by hyperglycemia,
aging, obesity, chronic inflammation, or autoimmunity, as ex-
amples, may supermodify these ligands, thus tipping the bal-
ance between “forms” likely to mediate rapid repair versus
those that engage the receptor chronically (Fig. 2). Indeed, we
predict that in simpler, monomeric forms, such as in uncom-
plicated and rapidly resolving acute stresses, these ligands
may activate RAGE and other innate receptors but that the
environment facilitates rapid removal of these ligands as repair
ensues. However, in chronically stressed environments, we
propose that these ligands may be more apt to achieve higher
order forms that cross the threshold to recognize RAGE
strongly and perhaps selectively. It is possible that aggregation
mechanisms may overwhelm natural clearance systems and
over-run the chronically injured environment and lead to sus-
tained recruitment and activity of RAGE.
In conclusion, striking the optimal balance in therapeutic
targeting of RAGE will require crossing a fine line of resolution
and repair between acute and chronic stimulation and thus,
resolution and repair or irreversible tissue injury. If our pre-
dictions are correct, then a chief challenge of ongoing work is
to identify the threshold for the ability of ligands to recruit
RAGE in distinct stresses. Once so identified, the optimal
pathways to targeting the receptor selectively in maladaptive
stress are likely to be uncovered.
ACKNOWLEDGMENTS
This work was supported by grants from the United States
Public Health Service and the Juvenile Diabetes Research
Foundation.
REFERENCES
1. Beisswenger, P. J., Howell, S. K., Nelson, R. G., Mauer, M., Szwergold,
B. S. (2003) -Oxoaldehyde metabolism and diabetic complications. Bio-
chem. Soc. Trans. 31, 1358 –1363.
2. Brownlee, M. (1996) Advanced glycation endproducts in diabetic compli-
cations. Curr. Opin. Endocrinol. Diabetes 3, 291–297.
3. Girones, X., Guuimera, A., Cruz-Sanchez, C. Z., Ortega, A., Sasaki, N.,
Makita, Z., Lafuente, J. V., Kalaria, R., Cruz-Sanchez, F. F. (2004) N ε
carboxymethyllysine in brain aging, diabetes mellitus and Alzheimer’s
disease. Free Radic. Biol. Med. 36, 1241–1247.
4. Ikeda, K., Higashi, T., Sano, H., Jinnouchi, Y., Yoshida, M., Araki, T.,
Ueda, S., Horiuchi, S. (1996) Carboxymethyllysine protein adduct is a
major immunological epitope in proteins modified with AGEs of the
Maillard reaction. Biochemistry 35, 8075– 8083.
5. Brown, B. E., Dean, R. T., Davies, M. J. (2005) Glycation of low density
lipoproteins by methylglyoxal and glycoaldehyde gives rise to the in vitro
formation of lipid-laden cells. Diabetologia 48, 361–369.
6. Wautier, M. P., Chappey, O., Corda, S., Stern, D. M., Schmidt, A. M.,
Wautier, J. L. (2001) Activation of NADPH oxidase by advanced glycation
endproducts (AGEs) links oxidant stress to altered gene expression via
RAGE. Am. J. Physiol. Endocrinol. Metab. 280, E685–E694.
7. Yan, S. D., Schmidt, A. M., Anderson, G. M., Zhang, J., Brett, J., Zou,
Y. S., Pinsky, D., Stern, D. (1994) Enhanced cellular oxidant stress by the
interaction of advanced glycation endproducts with their receptors/binding
proteins. J. Biol. Chem. 269, 9889 –9897.
8. Basta, G., Lazzerini, G., Del Turco, S., Ratto, G. M., Schmidt, A. M., De
Caterina, R. (2005) At least two distinct pathways generating reactive
oxygen species mediate vascular cell adhesion molecule-1 induction by
advanced glycation end products. Arterioscler. Thromb. Vasc. Biol. 25,
1401–1407.
9. Anderson, M. M., Requena, J. R., Crowley, J. R., Thorpe, S. R., Heinecke,
J. W. (1999) The myeloperoxidase system of human phagocytes generates
Nε-(carboxymethyl)lysine on proteins: a mechanism for producing ad-
vanced glycation end products at sites of inflammation. J. Clin. Invest.
104, 103–113.
10. Anderson, M. M., Heinecke, J. W. (2003) Production of N(ε)-(carboxy-
methyl)lysine is impaired in mice deficient in NADPH oxidase: a role for
phagocyte-derived oxidants in the formation of advanced glycation end-
products during inflammation. Diabetes 52, 2137–2143.
11. Kislinger, T., Fu, C., Huber, B., Qu, W., Taguchi, A., Yan, S. D.,
Hofmann, M., Yan, S. F., Pischetsrider, M., Stern, D., Schmidt, A. M.
(1999) Ne (carboxymethyl)lysine modifications of proteins are ligands for
RAGE that activate cell signaling pathways and modulate gene expres-
sion. J. Biol. Chem. 274, 31740 –31749.
Herold et al. RAGE, primal responses to stress and chronic disease 7
12. Oudes, A. J., Herr, C. M., Olsen, Y., Fleming, J. E. (1998) Age-dependent
accumulation of advanced glycation end-products in adult Drosophila
melanogaster.Mech. Ageing Dev. 100, 221–229.
13. Mironova, R., Niwa, T., Handzhiyski, Y., Sredovska, A., Ivanov, I. (2005)
Evidence for non-enzymatic glycosylation of Escherichia coli chromosomal
DNA. Mol. Microbiol. 55, 1801–1811.
14. Gomes, R. A., Miranda, H. V., Silva, M. S., Graca, G., Coelho, A. V.,
Ferreira, A. E., Cordeiro, C., Freire, A. P. (2006) Yeast protein glycation
in vivo by methylglyoxal. Molecular modification of glycolytic enzymes
and heat shock proteins. FEBS J. 273, 5273–5287.
15. Poggioli, S., Bakala, H., Friguet, B. (2002) Age-related increase of protein
glycation in peripheral blood lymphocytes is restricted to preferential
target proteins. Exp. Gerontol. 37, 1207–1215.
16. Poggioli, S., Mary, J., Bakala, H., Friguet, B. (2004) Evidence of prefer-
ential protein targets for age-related modifications in peripheral blood
lymphocytes. Ann. N. Y. Acad. Sci. 1019, 211–214.
17. Price, C. L., Sharp, P. S., North, M. E., Rainblow, S. J., Knight, S. C.
(2004) Advanced glycation end products modulate the maturation and
function of peripheral blood dendritic cells. Diabetes 53, 1452–1458.
18. Donato, R. (2001) S100: a multigenic family of calcium-modulated pro-
teins of the EF-hand type with intracellular and extracellular functional
roles. Int. J. Biochem. Cell Biol. 33, 637– 668.
19. Hofmann, M. A., Drury, S., Fu, C., Qu, W., Taguchi, A., Lu, Y., Avila, C.,
Kambham, N., Bierhaus, A., Nawroth, P., Neurath, M. F., Slattery, T.,
Beach, D., McClary, J., Nagashima, M., Morser, J., Stern, D., Schmidt,
A. M. (1999) RAGE mediates a novel proinflammatory axis: a central cell
surface receptor for S100/calgranulin polypeptides. Cell 97, 889 –901.
20. Huttunen, H. J., Kuja-Panula, J., Sorci, G., Agneletti, A. L., Donato, R.,
Rauvala, H. (2000) Coregulation of neurite outgrowth and cell survival by
amphoterin and S100 proteins through receptor for advanced glycation
end products (RAGE) activation. J. Biol. Chem. 275, 40096 40105.
21. Tang, D., Shi, Y., Kang, R., Li, T., Xiao, W., Wang, H., Xiao, X. (2007)
Hydrogen peroxide stimulates macrophages and monocytes to actively
release HMGB1. J. Leukoc. Biol. 81, 741–747.
22. Dumitriu, I. E., Bianchi, M. E., Bacci, M., Manfredi, A. A., Rovere-
Querini, P. (2007) The secretion of HMGB1 is required for the migration
of maturing dendritic cells. J. Leukoc. Biol. 81, 84 –91.
23. Hori, O., Brett, J., Slattery, T., Cao, R., Zhang, J., Chen, J., Nagashima, M.,
Nitecki, D., Morser, J., Stern, D., Schmidt, A. M. (1995) The receptor for
advanced glycation endproducts (RAGE) is a cellular binding site for
amphoterin: mediation of neurite outgrowth and co-expression of RAGE
and amphoterin in the developing nervous system. J. Biol. Chem. 270,
25752–25761.
24. Taguchi, A., Blood, D. C., del Toro, G., Canet, A., Lee, D. C., Qu, W.,
Tanji, N., Lu, Y., Lalla, E., Fu, C., Hofmann, M. A., Kislinger, T., Ingram,
M., Lu, A., Tanaka, H., Hori, O., Ogawa, S., Stern, D. M., Schmidt, A. M.
(2000) Blockade of amphoterin/RAGE signaling suppresses tumor growth
and metastases. Nature 405, 354 –360.
25. Wang, H., Bloom, O., Zhang, M., Vishnuhakat, J. M., Ombrellino, M., Che,
J., Frazier, A., Yang, H., Ivanova, S., Borovikova, L., Manogue, K. R.,
Faist, E., Abraham, E., Andersson, J., Andersson, U., Molina, P. E.,
Abumrad, N. N., Sama, A., Tracey, K. J. (1999) HMG-1 as a late mediator
of endotoxin lethality in mice. Science 285, 248 –251.
26. Yu, M., Wang, H., Ding, A., Golenbock, D. T., Latz, E., Czura, C. J.,
Fenton, M. J., Tracey, K. J., Yang, H. (2006) HMGB1 signals through
Toll-like receptor (TLR) 4 and TLR2. Shock 26, 174 –179.
27. Dumitriu, I. E., Baruah, P., Valentinis, B., Voll, R. E., Hermann, M.,
Nawroth, P. P., Arnold, B., Bianchi, M. E., Manfredi, A. A., Rovere-
Querini, P. (2005) Release of high mobility group box 1 by dendritic cells
controls T cell activation via the receptor for advanced glycation endprod-
ucts. J. Immunol. 174, 7506 –7515.
28. Chavakis, T., Bierhaus, A., Al-Fakhri, N., Schneider, D., Witte, S., Linn,
T., Nagashima, M., Morser, J., Arnold, B., Preissner, K. T., Nawroth, P. P.
(2003) The pattern recognition receptor (RAGE) is a counterreceptor for
leukocyte integrins: a novel pathway for inflammatory cell recruitment. J.
Exp. Med. 198, 1507–1515.
29. Orlova, V. V., Choi, E. Y., Xie, C., Chavakis, E., Bierhaus, A., Ihanus, E.,
Ballantyne, C. M., Gahmberg, C. G., Bianchi, M. E., Nawroth, P. P.,
Chavakis, T. (2007) A novel pathway of HMGB1-medaited inflammatory
cell recruitment that requires Mac-1 integrin. EMBO J. 26, 1129 –1139.
30. Yan, S. D., Chen, X., Fu, J., Chen, M., Zhu, H., Roher, A., Slattery, T.,
Nagashima, M., Morser, J., Migheli, A., Nawroth, P., Godman, G., Stern,
D., Schmidt, A. M. (1996) RAGE and amyloid-peptide neurotoxicity in
Alzheimer’s disease. Nature 382, 685– 691.
31. Yan, S. D., Zhu, H., Zhu, A., Golabek, A., Du, H., Roher, A., Yu, J., Soto,
C., Schmidt, A. M., Stern, D., Kindy, M. (2000) Receptor-dependent cell
stress and amyloid accumulation in systemic amyloidosis. Nat. Med. 6,
643– 651.
32. Ma, W., Lee, S. E., Guo, J., Qu, W., Hudson, B. I., Schmidt, A. M., Barile,
G. R. (2007) RAGE ligand upregulation of VEGF secretion in ARPE-19
cells. Invest. Ophthalmol. Vis. Sci. 48, 1355–1361.
33. Woltjer, R. L., Maezawa, I., Ou, J. J., Montine, K. S., Montine, T. J. (2003)
Advanced glycation endproduct precursor alters intracellular amyloid-/A
PP carboxy-terminal fragment aggregation and cytotoxicity. J. Alzhei-
mers Dis. 5, 467– 476.
34. Obrenovich, M. E., Monnier, V. M. (2004) Glycation stimulates amyloid
formation. Sci. Aging Knowledge Environ. 2004, pe3.
35. Miyata, T., Taneda, S., Kawai, R., Ueda, Y., Horiuchi, S., Hara, M.,
Maeda, K., Monnier, V. M. (1996) Identification of pentosidine as a native
structure for advanced glycation endproducts in -2 microglobulin con-
taining amyloid fibrils in patients with dialysis-related amyloidosis. Proc.
Natl. Acad. Sci. USA 93, 2353–2358.
36. Xie, J., Burz, D. S., He, W., Bronstein, I. B., Lednev, I., Shekhtman, A.
(2007) Hexameric calgranulin C (S100A12) binds to the receptor for
advanced glycated end products (RAGE) using symmetric hydrophobic
target-binding patches. J. Biol. Chem. 282, 4218 4231.
37. Lander, H. M., Tauras, J. M., Ogiste, J. S., Hori, O., Moss, R. A., Schmidt,
A. M. (1997) Activation of the receptor for advanced glycation endprod-
ucts triggers a p21(ras)-dependent mitogen-activated protein kinase path-
way regulated by oxidant stress. J. Biol. Chem. 272, 17810 –17814.
38. Schmidt, A. M., Hori, O., Chen, J. X., Li, J. F., Crandall, J., Zhang, J., Cao,
R., Yan, S. D., Brett, J., Stern, D. (1995) Advanced glycation endproducts
interacting with their endothelial receptor induce expression of vascular
cell adhesion molecule-1 (VCAM-1) in cultured human endothelial cells
and in mice. A potential mechanism for the accelerated vasculopathy of
diabetes. J. Clin. Invest. 96, 1395–1403.
39. Marsche, G., Semlitsch, M., Hammer, A., Frank, S., Weigle, B., Demling,
N., Schmidt, K., Windischhofer, W., Waeg, G., Sattler, W., Malle, E.
(2007) Hypochlorite-modified albumin colocalizes with RAGE in the
artery wall and promotes MCP-1 expression via the RAGE-ERK 1/2
MAP-kinase pathway. FASEB J. 21, 1145–1152.
40. Zeng, S., Feirt, N., Goldstein, M., Guarrera, J., Ippagunta, N., Ekong, U.,
Dun, H., Lu, Y., Qu, W., Schmidt, A. M., Emond, J. C. (2004) Blockade
of receptor for advanced glycation endproducts (RAGE) attenuates isch-
emia and reperfusion injury in the liver in mice. Hepatology 39, 422–
432.
41. Reddy, M. A., Li, S. L., Sahar, S., Kim, Y. S., Xu, Z. G., Lanting, L.,
Natarajan, R. (2006) Key role of Src kinase in S100b-induced activation
of the receptor for advanced glycation endproducts in vascular smooth
muscle cells. J. Biol. Chem. 281, 13685–13693.
42. Sakaguchi T., Yan, S. F., Yan, S. D., Rong, L. L., Sousa, M., Belov, D.,
Andrassy, M., Marso, S. P., Duda, S., Arnold, B., Liliensiek, B., Nawroth,
P. P., Stern, D. M., Schmidt, A. M., Naka, Y. (2003) Arterial restenosis:
central role of RAGE-dependent neointimal expansion. J. Clin. Invest.
111, 959 –972.
43. Bianchi, R., Adami, C., Giamanco, I., Donato, R. (2007) S100b binding to
RAGE in microglia stimulates COX-2 expression. J. Leukoc. Biol. 81,
108 –118.
44. Fukami, K., Ueda, S., Yamaghishi, S., Kato, S., Inagaki, Y., Takeuchi, M.,
Motomiya, Y., Bucala, R., Iida, S., Tamaki, K., Imaizumi, T., Cooper,
M. E., Okuda, S.. (2004) AGEs activate mesangial TGF--Smad signaling
via an angiotensin II type 1 receptor interaction. Kidney Int. 66, 2137–
2147.
45. Vincent, A. M., Perrone, L., Sullivan, K. A., Backus, C., Sastry, A. M.,
Lastoskie, C., Feldman, E. L. (2007) Receptor for advanced glycation end
products activation injures primary sensory neurons via oxidative stress.
Endocrinology 148, 548 –558.
46. Ishihara, K., Tsutsumi, K., Kawane, S., Nakajima, M., Kasaoka, T. (2003)
The receptor for advanced glycation endproducts (RAGE) directly binds to
ERK by a D-domain like docking site. FEBS Lett. 550, 107–113.
47. Sorci, G., Riuzzi, F., Agneletti, A. L., Marchetti, C., Donato, R. (2003)
S100b inhibits myogenic differentiation and myotube formation in a
RAGE-independent manner. Mol. Cell. Biol. 23, 4870 4881.
48. Horiuchi, S., Unno, Y., Usui, H., Shikata, K., Takaki, K., Koito, W.,
Sakamoto, Y., Nagai, R., Makino, K., Sasao, A., Wada, J., Makino, H.
(2005) Pathological roles of advanced glycation end product receptors
SR-1 and CD36. Ann. N. Y. Acad. Sci. 1043, 671– 675.
49. Hofmann, M. A., Drury, S., Hudson, B. I., Gleason, M. R., Qu, W., Lu, Y.,
Lalla, E., Chitnis, S., Monteiro, J., Stickland, M. H., Bucciarelli, L. G.,
Moser, B., Moxley, G., Itescu, S., Grant, P. J., Gregersen, P. K., Stern,
D. M., Schmidt, A. M. (2002) RAGE and arthritis: the G82S polymorphism
amplifies the inflammatory response. Genes Immun. 3, 123–135.
50. Liliensiek, B., Weigand, M. A., Bierhaus, A., Nicklas, W., Kasper, M.,
Hofer, S., Plachky, J., Grone, H. J., Kurschus, F. C., Schmidt, A. M., Yan,
S. D., Martin, E., Schleicher, E., Stern, D. M., Hammerling, G. G.,
Nawroth, P. P., Arnold, B. (2004) Receptor for advanced glycation end-
8 Journal of Leukocyte Biology Volume 82, August 2007 http://www.jleukbio.org
products (RAGE) regulates sepsis but not the adaptive immune response.
J. Clin. Invest. 113, 1641–1650.
51. Chen, Y., Yan, S. S., Colgan, J., Zhang, H. P., Luban, J., Schmidt, A. M.,
Stern, D., Herold, K. C. (2004) Blockade of late stages of autoimmune
diabetes by inhibition of the receptor for advanced glycation end products.
J. Immunol. 173, 1399 –1405.
52. Moser, B., Szabolcs, M. J., Ankersmit, H. J., Lu, Y., Qu, W., Weinberg, A.,
Herold, K. C., Schmidt, A. M. (2007) Blockade of RAGE suppresses
alloimmune reactions in vitro and delays allograft rejection in heart
transplantation. Am. J. Transplant. 7, 293–302.
53. Yan, S. S. D., Wu, Z. Y., Zhang, H. P., Furtado, G., Chen, X., Yan, S. F.,
Schmidt, A. M., Brown, C., Stern, A., LaFaille, J., Chess, L., Stern, D. M.,
Jiang, H. (2003) Suppression of experimental autoimmune encephalomy-
elitis by selective blockade of encephalitogenic T-cell infiltration of the
central nervous system. Nat. Med. 9, 287–293.
54. Moser, B., Desai, D. D., Chen, Y., Downie, M. P., Roth, G., Hudson, B. I.,
Szabolcs, M. J., Ankersmit, J. H., Herold, K., Schmidt, A. M., Clynes, R.
(2005) RAGE (receptor for advanced glycation endproducts) is required
for effective T cell priming in vivo. Circulation Suppl. II 112, 1596.
55. Cataldegirmen, G., Zeng, S., Feirt, N., Ippagunta, N., Dun, H., Qu, W., Lu,
Y., Rong, L. L., Hofmann, M. A., Kislinger, T., Pachydaki, S. I., Jenkins,
D. G., Weinberg, A., Lefkowitch, J., Rogiers, X., Yan, S. F., Schmidt,
A. M., Emond, J. (2005) RAGE limits regeneration after massive liver
injury by coordinated suppression of TNF-and NF-B. J. Exp. Med.
201, 473– 484.
56. Rong, L. L., Trojaborg, W., Qu, W., Kostov, K., Yan, S. D., Gooch, C.,
Szabolcs, M., Hays, A. P., Schmidt, A. M. (2004) Antagonism of RAGE
suppresses peripheral nerve regeneration. FASEB J. 18, 1812–1817.
57. Rong, L. L., Yan, S. F., Wendt, T., Hans-Wagner, D., Pachydaki, S.,
Bucciarelli, L. G., Adebayo, A., Qu, W., Lu, Y., Kostov, K., Lalla, E., Yan,
S. D., Gooch, C., Szabolcs, M., Trojaborg, W., Hays, A. P., Schmidt, A. M.
(2004) RAGE modulates peripheral nerve regeneration via recruitment of
both inflammatory and axonal outgrowth pathways. FASEB J. 18, 1818
1825.
58. Goova, M. T., Li, J., Kislinger, T., Qu, W., Lu, Y., Bucciarelli, L. G.,
Nowygrod, S., Wolf, B. M., Caliste, X., Yan, S. F., Stern, D. M., Schmidt,
A. M. (2001) Blockade of receptor for AGE (RAGE) restores effective
wound healing in diabetic mice. Am. J. Pathol. 159, 513–525.
59. Lalla, E., Lamster, I. B., Feit, M., Huang, L., Spessot, A., Qu, W.,
Kislinger, T., Lu, Y., Stern, D. M., Schmidt, A. M. (2000) Blockade of
RAGE suppresses periodontitis-associated alveolar bone loss in diabetic
mice. J. Clin. Invest. 105, 1117–1124.
Herold et al. RAGE, primal responses to stress and chronic disease 9
... It is known that macrophages take up circulating AGEs and induce the production of reactive oxygen species (ROS) and NF-κB signaling through activation of NADPH oxidase via RAGE. 10 Furthermore, AGE−RAGE interaction also induces the expression of TNF-α, a proinflammatory cytokine, and through positive feedback, promotes enhanced expression of RAGE and more of TNFα. Overall, under hyperglycaemic conditions, macrophages, specifically the M1 type (pro-inflammatory), play a major part in oxidative stress and promote diabetes-associated complications. ...
Article
Full-text available
Background: The inflammatory response in diabetes is strongly correlated with increasing amounts of advanced glycation end products (AGEs), methylglyoxal (MGO), aldosterone (Aldo), and activation of macrophages. Aldo is known to be associated with increased pro-inflammatory responses in general, but its significance in inflammatory responses under glycated circumstances has yet to be understood. In the current work, the aim of our study was to study the macrophage immune response in the presence of AGEs, MGO, and Aldo to comprehend their combined impact on diabetes-associated complications. Methods and Results: The viability of macrophages upon treatment with glycated HSA (Gly-HSA) promoted cell growth as the concentration increased from 100 to 500 μg/mL, whereas MGO at a high concentration (≥300 μM) significantly hampered cell growth. At lower concentrations (0.5–5 nM), Aldo strongly promoted cell growth, whereas at higher concentrations (50 nM), it was seen to inhibit growth when used for cell treatment for 24 h. Aldo had no effect on MGO-induced cell growth inhibition after 24 h of treatment. However, compared to MGO or Aldo treatment alone, an additional decrease in viability could be seen after 48 h of treatment with a combination of MGO and Aldo. Treatment with Aldo and MGO induced expression of TNF-α independently and when combined. However, when combined, Aldo and MGO significantly suppressed the expression of TGF-β. Aldo, Gly-HSA, and MGO strongly induced the transcription of NF-κB and RAGE mRNA and, as expected, also promoted the formation of reactive oxygen species. Also, by inducing iNOS and MHC-II and suppressing CD206 transcript expression, Gly-HSA strongly favored the differentiation of macrophages into M1 type (pro-inflammatory). On the other hand, the combination of Aldo and MGO strongly induced the expression of MHC-II, CD206, and ARG1 (M2 macrophage marker). These findings suggest that Gly-HSA, MGO, and Aldo differently influence macrophage survival, activation, and differentiation. Conclusions: Overall, this study gives an insight into the effects of glycated protein and MGO in the presence of Aldo on macrophage survival, activation, differentiation, and inflammatory response.
... The up-regulation of the expression of the receptor for advanced glycation end products (RAGE) occurs in response to the presence of its ligands, thereby establishing a positive feedback loop that amplifies its activation (Xie et al. 2013). Through neuroinflammatory mechanisms, the proinflammatory actions of the ligand-RAGE axis have been implicated in the pathophysiology of numerous diseases (Herold et al. 2007, Dong et al. 2022. Multiple studies examining RAGE levels in individuals with ASD suggest a link between the HMGB1/RAGE pathway and inflammation in ASD (Boso et al. 2006, Raouf et al. 2021. ...
... In addition, the inhibition of RAGE signaling by HMGB1 or RAGE antibodies protects against stroke-induced apoptosis, astrocyte swelling and reduced infarct volume according to some basic research [16][17][18][19]. A soluble form of RAGE (sRAGE) that is lacking the transmembrane and intracellular domains has been found to act as an inhibitor of RAGE by competing with canonical ligands, such as HMGB1, leading to the suppression of RAGE/NF-κB signaling cascades [20]. The potential therapeutic utility of sRAGE has attracted attention in terms of the prevention of RAGE-mediated disease pathogenesis. ...
Article
Full-text available
Impaired cerebral microcirculation after subarachnoid hemorrhage (SAH) has been shown to be related to delayed ischemic neurological deficits (DIND). We previously demonstrated the involvement of the receptor for advanced glycation end products (RAGE) in the pathogenesis of SAH related neuronal death. In the present study, we aimed to investigate the therapeutic effects of a recombinant soluble form of RAGE (sRAGE) on microcirculation impairment following SAH. Intrathecal injection of autologous blood in rats, mixed primary astrocyte and microglia cultures exposed to hemolysates and endothelial cells (ECs) from human brain microvascular exposed to glia-conditioned medium or SAH patient's CSF were used as experimental SAH models in vivo and in vitro. The results indicated that intrathecal administration of recombinant sRAGE significantly ameliorated the vasoconstriction of cortical arterioles and associated perfusion impairment, brain edema, reduced cell death, endothelial dysfunction, and improved motor performance at 24 and 48 h after SAH induction in rats. The in vitro results further showed that recombinant sRAGE significantly reduced astrocyte swelling and microglia activation, in parallel with decreased mRNA expression levels of pro-inflammatory cytokines including interleukin-6 (IL-6) and interleukin-1β (IL-1β) in vitro. Moreover, the in vitro model of SAH-induced p-eNOS and eNOS suppression, along with stress fiber formation in brain microvascular ECs, was effectively reversed by sRAGE treatment and led to a decrease in cleaved-caspase 3 expression. In summary, recombinant sRAGE effectively lessened microcirculation impairment and vascular injury after SAH via the mechanism of anti-inflammation, which may provide a potential therapeutic strategy for SAH.
... CML, CEL, and MGH-1-modified peptides bind the V domain of RAGE and induce intracellular signaling. However, other AGEs, such as pentosidine, do not induce any RAGE signaling [147,148]. A comprehensive investigation of the RAGE-activating capacity of the full spectrum of AGEs is urgently needed. ...
Article
Full-text available
Chronic obstructive pulmonary disease (COPD) is a disease of the airways and lungs due to an enhanced inflammatory response, commonly caused by cigarette smoking. Patients with COPD are often multimorbid, as they commonly suffer from multiple chronic (inflammatory) conditions. This intensifies the burden of individual diseases, negatively affects quality of life, and complicates disease management. COPD and comorbidities share genetic and lifestyle-related risk factors and pathobiological mechanisms, including chronic inflammation and oxidative stress. The receptor for advanced glycation end products (RAGE) is an important driver of chronic inflammation. Advanced glycation end products (AGEs) are RAGE ligands that accumulate due to aging, inflammation, oxidative stress, and carbohydrate metabolism. AGEs cause further inflammation and oxidative stress through RAGE, but also through RAGE-independent mechanisms. This review describes the complexity of RAGE signaling and the causes of AGE accumulation, followed by a comprehensive overview of alterations reported on AGEs and RAGE in COPD and in important co-morbidities. Furthermore, it describes the mechanisms by which AGEs and RAGE contribute to the pathophysiology of individual disease conditions and how they execute crosstalk between organ systems. A section on therapeutic strategies that target AGEs and RAGE and could alleviate patients from multimorbid conditions using single therapeutics concludes this review.
... Its main soluble form, sRAGE, is secreted into the alveolar space and is detectable in the serum, serving as a biomarker for the degree of lung injury [7,[12][13][14][15][16][17][18] and acting as a decoy receptor to downregulate the injurious pulmonary inflammatory response [19]. RAGE interacts with multiple ligands, including advanced glycation end-products (AGEs), high-mobility group box 1 protein (HMGB1), calgranulins/S100 proteins, amyloid peptides and macrophage adhesion ligand-1 (MAC-1) [20][21][22]. RAGE controls a variety of cellular processes such as cell proliferation and migration, inflammation, autophagy or apoptosis. Recent studies also point out a critical role for RAGE in modulating inflammation in macrophages [23], as cAMP elevation has been demonstrated to be capable of suppressing ligand-induced inflammatory reactions, including secretion of monocyte chemotactic protein 1 and macrophage recruitment, by promoting the conversion of RAGE isoforms from membrane RAGE to sRAGE [24]. ...
Article
Full-text available
The roles of thioredoxin-interacting protein (TXNIP) and receptor for advanced glycation end-products (RAGE)-dependent mechanisms of NOD-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome-driven macrophage activation during acute lung injury are underinvestigated. Cultured THP-1 macrophages were treated with a RAGE agonist (S100A12), with or without a RAGE antagonist; cytokine release and intracytoplasmic production of reactive oxygen species (ROS) were assessed in response to small interfering RNA knockdowns of TXNIP and NLRP3. Lung expressions of TXNIP and NLRP3 and alveolar levels of IL-1β and S100A12 were measured in mice after acid-induced lung injury, with or without administration of RAGE inhibitors. Alveolar macrophages from patients with acute respiratory distress syndrome and from mechanically ventilated controls were analyzed using fluorescence-activated cell sorting. In vitro, RAGE promoted cytokine release and ROS production in macrophages and upregulated NLRP3 and TXNIP mRNA expression in response to S100A12. TXNIP inhibition downregulated NLRP3 gene expression and RAGE-mediated release of IL-1β by macrophages in vitro. In vivo, RAGE, NLRP3 and TXNIP lung expressions were upregulated during experimental acute lung injury, a phenomenon being reversed by RAGE inhibition. The numbers of cells expressing RAGE, NLRP3 and TXNIP among a specific subpopulation of CD16+CD14+CD206- (“pro-inflammatory”) alveolar macrophages were higher in patients with lung injury. This study provides a novel proof-of-concept of complex RAGE–TXNIP–NLRP3 interactions during macrophage activation in acute lung injury.
... RAGE binds to a wide range of ligands, including AGEs, and certain S100/calgranulins, high mobility group Box1 (HMGB1), Mac-1, and amyloid-peptide, especially its oligomeric forms [34]. The production of inflammation and oxidative stress are two mechanisms by which AGEs can change the vasculature and enhance arterial stiffness [35]. ...
Article
Amidst several pathophysiological cascades, Advanced Glycation End products (AGEs) have been identified as a pivotal aetiology behind the pathogenesis and progression of cardiovascular disorders, by inducing oxidative stress and inflammation of myocardial and vascular tissues. Non-enzymatic glycation of reducing sugars with amino acids in proteins, lipids, and nucleic acids produce AGEs, which are a diverse set of compounds. Although AGEs are mostly generated endogenously, current research suggests that nutrition is a major exogenous source of AGEs. Extracellular and intracellular structure and function are affected by the presence and accumulation of AGEs in several cardiac cell types. AGEs give rise to several microvascular and macrovascular problems by establishing cross-links between molecules in the extracellular matrix's basement membrane as well as interacting with receptors for advanced glycation end products (RAGE). The transcription factor nuclear factor kappa B and its RAGE target genes are upregulated when RAGE is activated by AGEs. Engagement increases oxidative stress and triggers inflammatory and fibrotic responses, all of which contribute to the onset and progression of life-threatening cardiovascular diseases. This article discusses the probable targets of glycation in cardiac cells, as well as the underlying mechanisms that lead to heart failure.
... The membrane-bound RAGE (FL-RAGE) is a pattern recognition receptor that recognizes AGEs and several other ligands, some of which act as damage-associated molecular patterns (DAMPS) and pathogen-associated molecular patterns (PAMPs) [9][10][11]. Under physiological conditions, RAGE is expressed in the lungs while is almost undetectable in other tissues [12,13]; however, its levels are induced by ligand accumulation [14][15][16]. The cytoplasmic domain of RAGE binds to the formin DIAPH1 that is essential for RAGE/ligand-mediated activation and production of reactive oxygen species (ROS) eventually promoting inflammatory processes in ARDs [17,18]. ...
Article
Full-text available
Background: Advanced glycation end-products (AGEs) and their interaction with the receptor for advanced glycation end-products (RAGE) play a pivotal role in the development and progression of type 2 diabetes. In this retrospective cohort study, we explored the association of circulating levels of soluble RAGE (sRAGE) isoforms, i.e., endogenous secretory esRAGE and cleaved cRAGE, AGEs and their respective ratios with 15-year all-cause mortality in type 2 diabetes. Methods: Baseline AGEs and sRAGE isoforms concentration were measured by ELISA in 362 patients with type 2 diabetes and in 125 age- and gender-matched healthy control subjects (CTR). Independent predictors of mortality were determined using Cox proportional-hazards models and used to build and validate a nomogram for all-cause mortality prediction in type 2 diabetes. Results: AGEs, total sRAGE, cRAGE and the AGEs/sRAGE and AGEs/esRAGE ratios were significantly increased in patients with type 2 diabetes compared to CTR (p < 0.001). In CTR subjects, but not in type 2 diabetes patients, a significant negative correlation between cRAGE and age was confirmed (p = 0.003), whereas the AGEs/sRAGE (p = 0.032) and AGEs/cRAGE (p = 0.006) ratios were positively associated with age. At an average follow-up of 15 years (4,982 person-years), 130 deaths were observed. The increase in the AGEs/cRAGE ratio was accompanied by a higher risk of all-cause mortality in patients with type 2 diabetes (HR per each SD increment = 1.30, 95% CI 1.15-1.47; p < 0.001). Moreover, sRAGE was associated with the development of major adverse cardiovascular events (MACE) in type 2 diabetes patients without previous MACE (OR for each SD increase: 1.48, 95% CI 1.11-1.89). A nomogram based on age, sex, HbA1c, systolic blood pressure, and the AGEs/cRAGE ratio was built to predict 5-, 10- and 15-year survival in type 2 diabetes. Patients were categorized into quartiles of the monogram scores and Kaplan-Meier survival curves confirmed the prognostic accuracy of the model (log-rank p = 6.5 × 10- 13). Conclusions: The ratio between AGEs and the cRAGE isoform is predictive of 15-year survival in patients with type 2 diabetes. Our data support the assessment of circulating AGEs and soluble RAGE isoforms in patients with type 2 diabetes as predictors of MACE and all-cause mortality.
Article
Full-text available
The dysfunctional immune response and multiple organ injury in sepsis is a recurrent theme impacting prognosis and mortality, while the lung is the first organ invaded by sepsis. To systematically elucidate the transcriptomic changes in the main constituent cells of sepsis-injured lung tissue, we applied single-cell RNA sequencing to the lung tissue samples from septic and control mice and created a comprehensive cellular landscape with 25044 cells, including 11317 immune and 13727 non-immune cells. Sepsis alters the composition of all cellular compartments, particularly neutrophils, monocytes, T cells, endothelial, and fibroblasts populations. Our study firstly provides a single-cell view of cellular changes in septic lung injury. Furthermore, by integrating bulk sequencing data and single-cell data with the Scissors-method, we identified the cell subpopulations that are most associated with septic lung injury phenotype. The phenotypic-related cell subpopulations identified by Scissors-method were consistent with the cell subpopulations with significant composition changes. The function analysis of the differentially expressed genes (DEGs) and the cell-cell interaction analysis further reveal the important role of these phenotype-related subpopulations in septic lung injury. Our research provides a rich resource for understanding cellular changes and provides insights into the contributions of specific cell types to the biological processes that take place during sepsis-induced lung injury.
Article
The pathogenesis of obesity-related vascular disorders has not been fully elucidated. The fundamental role of inflammation in aging process is now widely recognized, particularly for atherosclerotic disease which begins before birth. The number of obese individuals worldwide has reached two billion, leading to an explosion of obesity-related vascular disorders associated with increased morbidity and mortality. Obesity, as a chronic low grade inflammatory process, is important risk factor for metabolic and cardiovascular disease. Despite a well-known genetic component, this risk appears to originate from several abnormalities in adipose tissue function associated with a chronic inflammatory state. In particular, obesity as the most common nutritional disorder in industrialized countries, is closely related to impaired endothelial function, a well-known marker of preatherosclerotic disease. These conditions disrupt vascular homeostasis by causing an imbalance between the nitric oxide pathway and the endothelin-1 system, with impaired insulin-stimulated endothelium-dependent vasodilation. Having in mind the growing population of overweight and obese people worldwide, along with an increasingly aging population, understanding the pathophysiology of obesity on cardiovascular system is essential. The mechanisms linking obesity-related vascular disorders and low grade inflammation in aging process are the focus of this paper.
Article
Full-text available
Attack by reactive oxygen intermediates, common to many kinds of cell/tissue injury, has been implicated in the development of diabetic and other vascular diseases. Such oxygen-free radicals can be generated by advanced glycation end products (AGEs), which are nonenzymatically glycated and oxidized proteins. Since cellular interactions of AGEs are mediated by specific cellular binding proteins, receptor for AGE (RAGE) and the lactoferrin-like polypeptide (LF-L), we tested the hypothesis that AGE ligands tethered to the complex of RAGE and LF-L could induce oxidant stress. AGE albumin or AGEs immunoisolated from diabetic plasma resulted in induction of endothelial cell (EC) oxidant stress, including the generation of thiobarbituric acid reactive substances (TBARS) and resulted in the activation of NF-kappaB, each of which was blocked by antibodies to AGE receptor polypeptides and by antioxidants. Infusion of AGE albumin into normal animals led to the appearance of malondialdehyde determinants in the vessel wall and increased TBARS in the tissues, activation of NF-kappaB, and induction of heme oxygenase mRNA. AGE-induced oxidant stress was inhibited by pretreatment of animals with either antibodies to the AGE receptor/binding proteins or antioxidants. These data indicate that interaction of AGEs with cellular targets, such as ECs, leads to oxidant stress resulting in changes in gene expression and other cellular properties, potentially contributing to the development of vascular lesions. Further studies will be required to dissect whether oxidant stress occurs on the cell surface or at an intracellular locus.
Article
Full-text available
Endotoxin, a constituent of Gram-negative bacteria, stimulates macrophages to release large quantities of tumor necrosis factor (TNF) and interleukin-1 (IL-1), which can precipitate tissue injury and lethal shock (endotoxemia). Antagonists of TNF and IL-1 have shown limited efficacy in clinical trials, possibly because these cytokines are early mediators in pathogenesis. Here a potential late mediator of lethality is identified and characterized in a mouse model. High mobility group–1 (HMG-1) protein was found to be released by cultured macrophages more than 8 hours after stimulation with endotoxin, TNF, or IL-1. Mice showed increased serum levels of HMG-1 from 8 to 32 hours after endotoxin exposure. Delayed administration of antibodies to HMG-1 attenuated endotoxin lethality in mice, and administration of HMG-1 itself was lethal. Septic patients who succumbed to infection had increased serum HMG-1 levels, suggesting that this protein warrants investigation as a therapeutic target.
Article
Full-text available
Protein glycation by methylglyoxal is a nonenzymatic post-translational modification whereby arginine and lysine side chains form a chemically heterogeneous group of advanced glycation end-products. Methylglyoxal-derived advanced glycation end-products are involved in pathologies such as diabetes and neurodegenerative diseases of the amyloid type. As methylglyoxal is produced nonenzymatically from dihydroxyacetone phosphate and d-glyceraldehyde 3-phosphate during glycolysis, its formation occurs in all living cells. Understanding methylglyoxal glycation in model systems will provide important clues regarding glycation prevention in higher organisms in the context of widespread human diseases. Using Saccharomyces cerevisiae cells with different glycation phenotypes and MALDI-TOF peptide mass fingerprints, we identified enolase 2 as the primary methylglyoxal glycation target in yeast. Two other glycolytic enzymes are also glycated, aldolase and phosphoglycerate mutase. Despite enolase's activity loss, in a glycation-dependent way, glycolytic flux and glycerol production remained unchanged. None of these enzymes has any effect on glycolytic flux, as evaluated by sensitivity analysis, showing that yeast glycolysis is a very robust metabolic pathway. Three heat shock proteins are also glycated, Hsp71/72 and Hsp26. For all glycated proteins, the nature and molecular location of some advanced glycation end-products were determined by MALDI-TOF. Yeast cells experienced selective pressure towards efficient use of d-glucose, with high methylglyoxal formation as a side effect. Glycation is a fact of life for these cells, and some glycolytic enzymes could be deployed to contain methylglyoxal that evades its enzymatic catabolism. Heat shock proteins may be involved in proteolytic processing (Hsp71/72) or protein salvaging (Hsp26).
Article
Full-text available
S100/calgranulin polypeptides are present at sites of inflammation, likely released by inflammatory cells targeted to such loci by a range of environmental cues. We report here that receptor for AGE (RAGE) is a central cell surface receptor for EN-RAGE (e xtracellular n ewly identified RAGE-binding protein) and related members of the S100/calgranulin superfamily. Interaction of EN-RAGEs with cellular RAGE on endothelium, mononuclear phagocytes, and lymphocytes triggers cellular activation, with generation of key proinflammatory mediators. Blockade of EN-RAGE/RAGE quenches delayed-type hypersensitivity and inflammatory colitis in murine models by arresting activation of central signaling pathways and expression of inflammatory gene mediators. These data highlight a novel paradigm in inflammation and identify roles for EN-RAGEs and RAGE in chronic cellular activation and tissue injury.
Article
The receptor for advanced glycation end products (RAGE), a newly-identified member of the immunoglobulin superfamily, mediates interactions of advanced glycation end product (AGE)-modified proteins with endothelium and other cell types. Survey of normal tissues demonstrated RAGE expression in situations in which accumulation of AGEs would be unexpected, leading to the hypothesis that under physiologic circumstances, RAGE might mediate interaction with ligands distinct from AGEs. Sequential chromatography of bovine lung extract identified polypeptides with M(r) values of approximate to 12,000 (p12) and approximate to 23,000 (p23) which bound RAGE. NH2-terminal and internal protein sequence data for p23 matched that reported previously for amphoterin. Amphoterin purified from rat brain or recombinant rat amphoterin bound to purified sRAGE in a saturable and dose-dependent manner, blocked by anti-RAGE IgG or a soluble form of RAGE (sRAGE). Cultured embryonic rat neurons, which express RAGE, displayed dose-dependent binding of I-125-amphoterin which was prevented by blockade of RAGE using antibody to the receptor or excess soluble receptor (sRAGE). A functional correlate of RAGE-amphoterin interaction was inhibition by anti-RAGE F(ab')(2) and sRAGE of neurite formation by cortical neurons specifically on amphoterin-coated substrates. Consistent with a potential role for RAGE-amphoterin interaction in development, amphoterin and RAGE mRNA/antigen were co-localized in developing rat brain. These data indicate that RAGE has physiologically relevant ligands distinct from AGEs which are likely, via their interaction with the receptor, to participate in physiologic processes outside of the context of diabetes and accumulation of AGEs.