You are on page 1of 12

Pharmacology & Therapeutics 177 (2017) 44–55

Contents lists available at ScienceDirect

Pharmacology & Therapeutics

journal homepage: www.elsevier.com/locate/pharmthera

Associate editor: B.J. Song

Advanced glycation end-products produced systemically and by


macrophages: A common contributor to inflammation and
degenerative diseases
Kyunghee Byun a,b,1, YongCheol Yoo c,1, Myeongjoo Son b, Jaesuk Lee a, Goo-Bo Jeong b, Young Mok Park c,⁎,1,
Ghasem Hosseini Salekdeh d,⁎⁎,1, Bonghee Lee a,b,⁎⁎⁎,1
a
Center for Genomics and Proteomics, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 406-840, Republic of Korea
b
Department of Anatomy and Cell Biology, Gachon University Graduate School of Medicine, Incheon 406-799, Republic of Korea
c
Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 305-811, Republic of Korea
d
Department of Molecular Systems Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran

a r t i c l e i n f o a b s t r a c t

Available online 13 February 2017 Advanced glycation end products (AGEs) and their receptor have been implicated in the progressions of many in-
tractable diseases, such as diabetes and atherosclerosis, and are also critical for pathologic changes in chronic degen-
Keywords: erative diseases, such as Alzheimer's disease, Parkinson's disease, and alcoholic brain damage. Recently activated
Advanced glycation end products (AGEs) macrophages were found to be a source of AGEs, and the most abundant form of AGEs, AGE-albumin excreted by
Receptor for AGEs (RAGE)
macrophages has been implicated in these diseases and to act through common pathways. AGEs inhibition has
Macrophage
been shown to prevent the pathogenesis of AGEs-related diseases in human, and therapeutic advances have result-
Inflammation
Degenerative diseases
ed in several agents that prevent their adverse effects. Recently, anti-inflammatory molecules that inhibit AGEs
have been shown to be good candidates for ameliorating diabetic complications as well as degenerative diseases.
This review was undertaken to present, discuss, and clarify current understanding regarding AGEs formation in as-
sociation with macrophages, different diseases, therapeutic and diagnostic strategy and links with RAGE inhibition.
© 2017 The Author(s). Published by Elsevier Inc. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).

Abbreviations: AD, Alzheimer's disease; ADMA, asymmetric dimethylarginine; AGEs, advanced glycation end products; AFLD, alcoholic fatty liver disease; ALI, acute lung injury; ALS,
amyotrophic lateral sclerosis; AMI, acute myocardial infarction; APP, acute phase protein; Aβ, amyloid beta; Bax, BCL2 associated X, apoptosis regulator; CaMKKβ, calcium/calmodulin-
dependent protein kinase kinase beta; CCL3, chemokine (C\ \C motif) ligand 3; CCL4, chemokine (C\ \C motif) ligand 4; CD36, cluster of differentiation 36; CKD, chronic kidney disease;
CMA, N(omega)-carboxymethylarginine; CML, N(6)-carboxymethyllysine; COPD, chronic obstructive pulmonary disease; CTGF, connective tissue growth factor; CVDs, cardiovascular
diseases; DAMPs, damage-associated molecular pattern molecules; DN, diabetic nephropathy; EGCG, epigallocatechin gallate; Egr-1, early growth response-1; ERK1/2, extracellular
signal-regulated kinase 1/2; FEEL-1, fasciclin EGF-like, laminin-type EGF-like, and link domain-containing scavenger receptor-1; FEEL-2, fasciclin EGF-like, laminin-type EGF-like, and
link domain-containing scavenger receptor-2; HDL, high-density lipoprotein; HMGB1, high mobility group protein B1; HUVEC, human umbilical vein endothelial cell; IL-6, interleukin-
6; IL-10, interleukin-10; IL-1β, interleukin-1 beta; INF-γ, interferon gamma; IPF, idiopathic pulmonary fibrosis; JAK, janus kinase; JNK, c-Jun N-terminal kinase; Lox-1, lectin-like
oxidized low-density lipoprotein receptor 1; LPS, lipopolysaccharide; Mac-1, macrophage-1 antigen; MAPK, mitogen activated protein kinase; MCP-1, monocyte chemoattractant
protein-1; MMP, matrix metalloproteinase; MMP-13, matrix metalloproteinase 13; NADPH, nicotinamide adenine dinucleotide phosphate; NF-κB, nuclear factor kappa B; NO, nitric
oxide; NOS, nitric oxide synthases; OA, osteoarthritis; PAR, protease-activated receptor; PAR1, protease-activated receptor 1; PAR2, protease-activated receptor 2; PAR3, protease-
activated receptor 3; PAR4, protease-activated receptor 4; PD, Parkinson's disease; PDGF, platelet-derived growth factor; PG, prostaglandin; PGE2, prostaglandin E2; PPAR-α,
peroxisome proliferator activated receptor-alpha; PPAR-γ, peroxisome proliferator-activated receptor gamma; RA, rheumatoid arthritis; Rac2, ras-related C3 botulinum toxin substrate
2; RAGE, receptor for advanced glycation end-products; ROS, reactive oxygen species; S100A8, S100 calcium binding protein A8; S100A9, S100 calcium binding protein A9; S100A12,
S100 calcium binding protein A12; SAPK, stress-activated protein kinase; SCI, spinal cord injury; sRAGE, soluble RAGE; SR-BI, scavenger receptor class B type I; SR-BII, scavenger
receptor class B type II; STAT, signal transducer and activator of transcription; STAT1, signal transducer and activator of transcription 1; STAT3, signal transducer and activator of
transcription 3; TGF-β, transforming growth factor beta; TNF, tumor necrosis factor; TNF-α, tumor necrosis factor alpha; VEGF, vascular endothelial growth factor.
⁎ Correspondence to: Y.M. Park, Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34047, Republic of Korea.
⁎⁎ Correspondence to: G.H. Salekdeh, Department of Molecular Systems Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
⁎⁎⁎ Correspondence to: B. Lee, Center for Genomics and Proteomics, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea.
E-mail addresses: ympark@kbsi.re.kr (Y.M. Park), Salekdeh@royaninstitute.org (G.H. Salekdeh), bhlee@gachon.ac.kr (B. Lee).
1
These authors contributed equally to this work.

http://dx.doi.org/10.1016/j.pharmthera.2017.02.030
0163-7258/© 2017 The Author(s). Published by Elsevier Inc. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
K. Byun et al. / Pharmacology & Therapeutics 177 (2017) 44–55 45

Contents

1. Introduction. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 45
2. Advanced glycation end-products (AGEs) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 45
3. Receptors for AGEs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 45
4. Mediators of macrophage activation and AGE formation . . . . . . . . . . . . . . . . . . . . . . . . . . . 45
47
5. Development of therapeutics against AGEs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 45
49
6. Conclusion and future perspectives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 45
50
Conflict of interest statement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 47
51
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 47
51
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 51

1. Introduction and oxidative stress (Takeuchi & Yamagishi, 2004). AGEs accumulate
in the extra cellular matrix of various tissues and critically contribute
Advanced glycation end products (AGEs) have long been considered to chronic diseases (Bierhaus et al., 1998; Tessier et al., 1999). Further-
potent toxic molecules that promote host cell death and contributing to more, AGEs have been shown to become antigenic, and thus, to induce
organ damage in man. AGEs can induce the development or progression immune responses (Ge et al., 2005; Kurien & Scofield, 2008). AGEs also
of not only diabetic complications but the pathophysiologies of many have adverse effects on cellular functions by cross-linking intracellular
diseases, including cardiovascular diseases (CVDs) and neurodegenera- and extracellular proteins, which trigger diverse diseases, such as arthri-
tive diseases like Alzheimer's disease (AD), Parkinson's disease (PD), tis, neurological disorders, cancers, diabetes and cardiovascular disor-
and alcoholic brain damage (Bayarsaikhan et al., 2016; Byun et al., ders, (Abe et al., 2004; Jono et al., 2002; Russo & Frangogiannis, 2016;
2012a, 2014; Simm et al., 2007; Srikanth et al., 2011; Yan et al., 2007). Ryan et al., 2014) (Fig. 1).
AGEs bind with their receptor RAGE (receptor for advanced glycation
end products), which is highly expressed during host cell death through 2.1. Intracellular formation and the toxic properties of AGEs
reactive oxygen species (ROS) and strong activations of extracellular
signal-regulated kinase 1/2 (ERK1/2) ERK1/2 and nuclear factor kappa 2.1.1. Formation of AGEs
B (NF-κB) pathways in many diseases. (Teismann et al., 2012; Wang AGEs are formed by glycation, which is a nonenzymatic reaction be-
et al., 2013, 2014). Inhibition of AGEs from blood or activated macro- tween ketones or aldehydes and the amino groups of various proteins
phages had been well shown to decrease the pathogenesis of several and contributes to protein aging (Gkogkolou & Böhm, 2012;
diseases and several agents have been developed to ameliorate their ad- Ramasamy et al., 2012). Protein glycation occurs in both normal and hy-
verse effects. Anti-inflammatory molecules that inhibit AGEs may be perglycemic condition, and forms irreversible AGEs. This process begins
good candidate treatments for diabetic complications and degenerative with the conversion of reversible Schiff base adducts to more stable, co-
diseases. In this review, we discuss the formation of AGEs and RAGE, valently-bound Amadori rearrangement products and finally the
their association with the mononuclear phagocytic system in diseases, Maillard reaction (Ansari & Dash, 2013; Ott et al., 2014). AGEs are also
the current therapeutic agents used and their adverse effects, and finally formed in the absence of hyperglycemia by homeostatic imbalances,
we suggest a strategy for targeting macrophage AGEs and RAGE interac- such as redox imbalances (Limongi & Baldelli, 2016), aging
tion for possible drug target. (Karumanchi et al., 2015), kidney disease (Arsov et al., 2014), or auto-
immune diseases (Nienhuis et al., 2009). In addition, AGEs are derived
2. Advanced glycation end-products (AGEs) from foods (e.g. milk, meat, coffee, cheese), especially those prepared
under high temperature conditions, stored for long periods, or with
AGEs are a heterogeneous group of irreversible products resulting food additives (Luevano-Contreras & Chapman-Novakofski, 2010).
from nonenzymatic glycation and oxidation of proteins, nucleic acids,
and lipids (Goldin et al., 2006; Singh et al., 2001). Glycation produces 2.1.2. Toxic properties of AGEs
AGEs compounds with toxic properties associated with inflammation Elevated levels of AGEs support the formations of reactive oxygen
and nitrogen species which in turn induce further AGEs formation.
AGEs induce oxidative stress by activating RAGE, which results in mito-
chondrial dysfunction (Lane et al., 2015; Ward et al., 2013). Under oxi-
dative stress, mitochondrial Ca2 + accumulation leads to a cell death
signal (Mahali et al., 2011), and AGEs excreted by activated macro-
phages also induce mitochondrial dysfunction and cell death. Extensive
studies have demonstrated that mitochondrial dysfunction is an impor-
tant factor in the pathogenesis of AD (Aliev et al., 2010), alcoholic fatty
liver disease (AFLD) (Song et al., 2014), diabetes (Rolo & Palmeira,
2006), and chondrocyte degeneration (Blanco et al., 2004) (Fig. 2).

3. Receptors for AGEs

AGEs can interact with two types of cell surface receptors. Scavenger
receptors are predominantly involved in AGEs capture, removal and
degradation. This group of receptors includes type I and type II macro-
phage scavenger receptors, cluster of differentiation 36 (CD36), fasciclin
EGF-like, laminin-type EGF-like, and link domain-containing scavenger
Fig. 1. Association of advanced glycation end products (AGEs) and human diseases. AGEs
are related to disease incidence of several organs through common mechanism of
receptor-1 (FEEL-1) and fasciclin EGF-like, laminin-type EGF-like,
oxidative stress, neovascularization, inflammation or cross linking with extracellular and link domain-containing scavenger receptor-2 (FEEL-2), scavenger
matrix (ECM). receptor class B type I (SR-BI) and scavenger receptor class B type II
46 K. Byun et al. / Pharmacology & Therapeutics 177 (2017) 44–55

Fig. 2. AGEs from activated macrophage and its receptor mediated tissue injury via mitochondrial dysfunction. AGE is one of major five protein modification induced by ROS/RNS after
exposure to alcohol, high fat diet, tobacco, drugs or gene alterations. AGEs induce RAGE increase and mitochondrial dysfunction leading to tissue injury.

(SR-BII), and lectin-like oxidized low-density lipoprotein receptor 1 2007), Aβ (Stern et al., 2002), amphoterin, and calgranulins, and then
(Lox-1) (Miyazaki et al., 2002; Ott et al., 2014). The other type of AGEs activate several condition-dependent cellular pathways (Li et al.,
receptor, RAGE initiates specific cellular signaling events in response 2004). RAGE is expressed in various tissues including brain, heart,
to AGEs exposure. RAGE is a member of immunoglobulin (Ig) superfam- liver, lung, kidney, and cartilage and it plays an important role in disease
ily, and binds multi-ligands like AGEs (Huttunen et al., 1999), high mo- progression. Expression level of RAGE is minimal under normal condi-
bility group protein B1 (HMGB1) (Dhumale et al., 2015), S100 (Donato, tions but increases in disease states, such as AD, PD, alcoholic brain

Fig. 3. Cell specific ligands for Receptor for AGEs (RAGE) and their high-lighted signaling pathways in relevant diseases. AD; Alzheimer's disease, PD; Parkinson's disease, Alc; Alcoholic
brain damage, AMI; Acute myocardial infarction, LC; Liver cirrhosis, IPF; Idiopathic pulmonary fibrosis, DN; diabetic nephropathy, OA; osteoarthritis, F; fibrosis, I; inflammation, O;
oxidative stress, A; autophagy, D; degeneration, Cd; cell death.
K. Byun et al. / Pharmacology & Therapeutics 177 (2017) 44–55 47

damage, vascular disease, diabetes, acute myocardial infarction (AMI), of HMGB1 binding to RAGE provokes inflammatory reactions via
chronic kidney disease (CKD), hepatic fibrosis, chronic obstructive pul- ERK1/2-NF-kB signaling, which turn on transcription of TNF-α, interleu-
monary disease (COPD), and inflammatory disease, which may be the kin-6 (IL-6), chemokine (C\\C motif) ligand 3 (CCL3), chemokine (C\\C
result of cell damage caused by the overproductions of ROS, cytokines, motif) ligand 4 (CCL4) and C-X-C motif chemokine 12 (CXCL12) in DN
and pro-inflammatory molecules (Fig. 3, Supplementary Table 1). and enhances asymmetric dimethylarginine (ADMA) in CKD
(Nakamura et al., 2009).
3.1. Intracellular signaling pathway of RAGE in disease
3.1.6. Chondrocytes and RAGE
AGEs dependent RAGE expression is well documented in many dis- The binding AGEs to RAGE activate the JNK/p38, peroxisome
eases. The signaling pathways downstream of RAGE are similar and proliferator-activated receptors alpha (PPAR-α), and NF-κB signaling
eventually lead to mitochondrial dysfunction and cell death. The de- pathway and these signaling pathways associated with inflammatory
tailed summary is provided below and in Fig. 3 and Supplementary and oxidative responses in osteoarthritis (OA). Furthermore, high levels
Table 1. of HMGB1 have been reported in the synovial fluid of arthritis patients
(Kokkola et al., 2002), and the interaction between HMGB1 and RAGE
3.1.1. Relations between RAGE and neuronal death induces the expressions of TNF-α and interleukin 1 beta (IL-1β) on
RAGE is highly expressed on neurons during neurodegenerative pro- chondrocytes (Terada et al., 2011). Furthermore, when the expression
cesses, such as AD, PD, stroke, alcoholic brain damage, and amyotrophic of S100 calcium binding protein A12 (S100A12) and A4 (S100A4) is in-
lateral sclerosis (ALS). In the presence of inflammation, binding of AGEs creased and it stimulates inflammation and degeneration via the RAGE
to RAGE strongly induces the activations of the NF-κB pathways mediated MAPK, NF-κB and MMP-13/vascular endothelial growth fac-
(Teismann et al., 2012; Wang et al., 2013, 2014). Furthermore, the inter- tor (VEGF)/p38 and NF-κB signaling pathway in OA (Nakashima et al.,
action between AGE-albumin and RAGE induces neuronal cell death 2012; Yammani et al., 2009).
through stress-activated protein kinase (SAPK)/c-Jun N-terminal kinase
(JNK)/BCL2 associated X, apoptosis regulator (Bax) in AD (Byun et al., 4. Mediators of macrophage activation and AGE formation
2012a) and the SAPK/JNK/p38 pathway in the alcoholic brain damage
(Byun et al., 2014). Furthermore, increased HMGB1 binding with The macrophages originate from bone marrow and then migrate and
RAGE can cause neuronal cell death through NF-κB activation in stroke circulate in different tissues, such as the brain, liver, heart, kidney, and
and AD (Bortolotto & Grilli, 2016; Wang et al., 2010). S100 protein in- cartilage (Kraft & Harry, 2011; Varol et al., 2015). Activated macro-
duces neuronal cell death through the NF-κB/tumor necrosis factor phages often induce inflammatory degeneration of host cells or dys-
alpha (TNF-α) pathway in PD (Abdelsalam & Safar, 2015; Sathe et al., function in AD, PD, stroke, vascular disease, and inflammatory
2012), and in AD, binding of Aβ to RAGE also leads to neurodegenera- diseases. Macrophages are activated by exposure to various stimuli,
tion and neuronal cell death through p21ras, ERK1/2, p38, SAPK/JNK, such as cytokines, proteases, prostaglandins (PGs), nitric oxide (NO),
phosphoinositol-3 kinase, and the janus kinase (JAK)/signal transducer free radicals or RAGE ligands, including Aβ, AGEs, HMGB1 and lipopoly-
and activator of transcription (STAT) pathway (Origlia et al., 2010; Son saccharide (LPS), under pro- and inflammatory conditions (Fig. 4).
et al., 2012).
4.1. Cytokines
3.1.2. Cardiomyocytes and RAGE
RAGE expression of cardiomyocytes is increased in acute myocardial Cytokines are involved in systemic inflammation and degenerative
infarction by AGEs, HMGB1 and S100, and these proteins induce cell diseases. They can be released by neurons, cardiomyocytes, hepato-
death via activation of the MAPK pathway (Aleshin et al., 2008; cytes, alveolar epithelial cells, neutrophils, eosinophils, or mast cells at
Andrassy et al., 2008; Tsoporis et al., 2010). various levels when cells are stimulated or in the presence of disease
conditions. These cytokines such as α-synuclein (Kim & Joh, 2006),
3.1.3. Hepatocytes and RAGE Chromogranin A (Heneka et al., 2010, and Interferon gamma (INF-γ)
Binding of AGEs to RAGE strongly induces the phosphorylation of (Gensel & Zhang, 2015) were well reported as activating macrophages
uncoupling protein-2 (UCP-2) and ATP synthesis (Kuhla et al., 2011). under disease conditions.
HMGB1 released from damaged hepatocyte after injury interacts with
RAGE and leads to hepatic fibrosis by activating SMAD2 and phosphor- 4.2. Proteases
ylating ERK1/2 (Kao et al., 2014) or JNK/ERK1/2 during early growth re-
sponse-1 (Egr-1) (Zeng et al., 2009). Serine proteases cleave their receptors to modulate important bio-
logical processes including host defense, chemotaxis, cytokine and
3.1.4. Alveolar epithelial cells and RAGE growth factor release, vascular function, tissue repair, apoptosis, inflam-
The RAGE binding with AGEs and HMGB1 has been well described in mation and immune response (Steinhoff et al., 2005). Protease-activat-
idiopathic pulmonary fibrosis (IPF). Binding of AGEs and RAGE inhibits ed receptors (PARs) compose a subfamily of G protein-coupled
TGF-β induced ERK1/2 and SMAD phosphorylation (Song et al., 2011) receptors with seven transmembrane receptors that couple to guano-
and up-regulates Egr-1, which also activates RAGE transcription in sine-nucleotide-binding proteins (Brass & Molino, 1997). The PAR fam-
COPD (Reynolds et al., 2008b). In addition, the interaction between ily has four members, namely, protease-activated receptor 1 (PAR1),
HMGB1 and RAGE leads to alveolar disruption and fibrosis due to the ac- protease-activated receptor 2 (PAR2), protease-activated receptor 3
tivation of TGF-β via the productions of platelet-derived growth factor (PAR3), and protease-activated receptor 4 (PAR4), and of these, PAR2
(PDGF) in IPF (He et al., 2007). can modulate the activities of various cells, including epithelial cells,
smooth muscle cells, endothelial cells, and macrophages in a variety of
3.1.5. Renal tubular epithelial cells and RAGE tissues (Nystedt et al., 1996; Steinhoff et al., 2005).
Interaction between AGEs and RAGE stimulates oxidative stress and
promotes diabetic nephropathy (DN) progression. Importantly, AGE- 4.3. Prostaglandins (PGs)
RAGE acts to the mitogen activated protein kinase (MAPK)/NF-κB and
protein kinase C signaling pathway (Yamagishi & Matsui, 2010) or pro- PGs act as critical regulators during the initiation of inflammatory
mote to connective tissue growth factor (CTGF) and TGF-β during in- processes. PG expressions are significantly increased in infected tissues,
flammation and fibrotic process (Cheng et al., 2015a). Up-regulation and contribute to the development of the principal signs of the acute
48 K. Byun et al. / Pharmacology & Therapeutics 177 (2017) 44–55

Fig. 4. Activators of macrophages and secreted proteins from activated macrophages. RAGE ligands also induce activation of macrophages and they are secreted from activated
macrophages. AGEs; advanced glycation end products, HMGB1; high-mobility group protein B1, Mac-1; macrophage-1 antigen, LPS; lipopolysaccharide, Aβ; amyloid beta, IL-10;
interleukin-10, NO; nitric oxide, ROS; reactive oxygen species.

inflammatory phase (Ricciotti & FitzGerald, 2011). Prostaglandin E2 4.6. RAGE ligands
(PGE2) is one of the most abundant PGs and exhibits variable biological
activities, which include the regulations of blood pressure, gastrointes- RAGE ligands including Aβ, AGEs, HMGB1, LPS, macrophage-1 anti-
tinal integrity, and immune response (Legler et al., 2010). gen (Mac-1), phosphatidylserine and S100 also modulate important bi-
ological processes required for macrophage activation (Chavakis et al.,
2004; He et al., 2011; Hofmann et al., 1999; Schmidt et al., 2001). The
4.4. Acute phase proteins (APPs) various types of RAGE ligands that induce macrophage activation in de-
generative diseases are summarized in Table 1. Aβ is cleaved from am-
APPs response to inflammation may be an increase (positive acute- yloid precursor protein and is the main cause of AD, a major component
phase proteins, APPs) or decrease (negative APPs) in plasma concentra- of amyloid plaque (Fang et al., 2010). Nuclear HMGB1 regulates pro-in-
tion (Gruys, Toussaint, Niewold, & Koopmans, 2005). Positive APPs are flammatory factors, and HMGB1 can be released by neurons in the dam-
involved in different physiological functions in the immune system. aged brain (Fang et al., 2010). HMGB1 up-regulation leads to microglial
For example, haptoglobin acts to inhibit or prevent microbe growth activation in many diseases including alcoholic brain damage (Crews &
and alpha 2 macroglobulin and serpins provide negative feedback on Vetreno, 2014, 2016), stroke (Liesz et al., 2015; Xiong et al., 2016), spi-
the inflammatory processes (Jain, Gautam, & Naseem, 2011). nal cord injury (SCI) (Kikuchi et al., 2011), AMI (Herzog et al., 2014) and
Some of these proteins act to inhibit or prevent microbe growth, acute lung injury (ALI) (Deng et al., 2013). LPS can stimulate macro-
while others modulate negative feedback from inflammatory processes phages and monocytes to secrete various inflammatory mediators, in-
which include albumin, transferrin, transthyretin, transcortin, retinol- cluding IL-6 (Hirohashi & Morrison, 1996). LPS also induces
binding protein and anti-thrombin (Gruys et al., 2005; Ritchie et al., macrophage activation in SCI (Xie et al., 2004), hypertension (Hernanz
1999). et al., 2004), ALI (Zhu et al., 2013), liver cirrhosis (Jirillo et al., 2002),
IPF (Freeburn et al., 2005), and COPD (Miles et al., 1999). S100 members
4.5. Nitric oxide and free radicals also activate macrophages, and are also secreted by activated macro-
phages (Donato, 1999).
4.5.1. Nitric oxide (NO)
NO is a free radical and an important cellular signaling mediator re-
Table 1
lated to various pathological and physiological processes, including the
RAGE ligands that activate macrophages under inflammatory conditions and in degener-
modulation of blood pressure, neurotransmission, and of macrophage ative diseases.
cytotoxicity. NO is synthesized by nitric oxide synthase (NOS) from ox-
Cytokine Disease Reference
ygen and NADPH as cofactor of NOS (Mayer et al., 1990). NO is well
known as endogenous mediator during vasodilator inflammatory pro- HMGB1 Alcoholic brain damage Crews and Vetreno (2014)
cesses (Wallace, 2005). Crews and Vetreno (2016)
Spinal cord injury Kikuchi et al. (2011)
Stroke Liesz et al. (2015)
Xiong, Liu, and Yang (2016)
4.5.2. Oxidative stress Acute myocardial injury Herzog et al. (2014)
In oxidative stress, greater amounts of oxygen free radicals are found S100A8/9 Osteoarthritis van Lent et al. (2012)
under disease states or after exposure to toxic agents and are increased Aβ Alzheimer's disease Fang et al. (2010)
by environmental stresses such as UV or heat. Oxidative stress elevates LPS Spinal cord injury Xie, Smith, and Van Eldik (2004)
Hypertension Hernanz, Briones, Alonso, Vila, and
the expressions of inflammatory mediators, which induce macrophage
Salaices (2004)
activation in the presence of inflammation (Macdonald, Galley, & Liver cirrhosis Jirillo et al. (2002)
Webster, 2003), AMI (Di Filippo, Cuzzocrea, Rossi, Marfella, & Chronic obstructive Miles, Bowman, Rao, Baatz, and
D'Amico, 2006), COPD (Rahman, 2005), OA (Ziskoven et al., 2010) and pulmonary disease Huffman (1999)
neurodegenerative disease, such as AD (Zhu et al., 2004), and PD Idiopathic pulmonary fibrosis Freeburn, Armstrong, and Millar (2005)
Acute lung injury Zhu et al. (2013)
(Dias, Junn, & Mouradian, 2013).
K. Byun et al. / Pharmacology & Therapeutics 177 (2017) 44–55 49

4.7. Proteomic analysis of activated macrophages Aβ interacts with AGEs and induces microglial activation and neuroin-
flammation via ERK1/2 and NF-κB activation (Lv et al., 2014). Several
To further understand the molecular mechanism responsible for authors have demonstrated AGEs accumulation in the brains of patients
macrophage activation, proteomic studies have been recently conduct- with AD (Ahn et al., 2008) or alcoholic brain damage (Byun et al., 2014).
ed in a variety of disease models, including neurodegenerative disease In addition, Aβ stimulated human microglial cells significantly synthe-
(Ma et al., 2013; Reynolds et al., 2008a; Yoo et al., 2015; Zhou et al., size and release more albumin (Ahn et al., 2008), and abundant quanti-
2005), acute lung injury/acute respiratory distress syndrome (Dong et ties of AGE-albumin, a new RAGE ligand (Byun et al., 2012a). Aβ plaques
al., 2013), kidney stone disease (Singhto et al., 2013), metabolic diseases are co-localized with AGE-albumin in Aβ1-42 injected rat and human
(Freiwald et al., 2013; Kratz et al., 2014), cancer (Kobayashi et al., 2009; AD brain (Byun et al., 2012b) and the AGE-albumin induces calcium in-
Zhu et al., 2015), atherosclerosis (Burillo et al., 2009; Kang et al., 2006), flux via up-regulation of RAGE, and this leads to neuronal death in AD
and virus infections (Lietzén et al., 2011; Miettinen et al., 2012). The (Byun et al., 2012a). Furthermore, alcohol-stimulated microglial cells
proteins secreted by activated macrophages are being actively investi- also exhibit much increased synthesis and release of AGE-albumin and
gated in terms of their abilities to damage or protect tissues. induce neuronal cell death (Byun et al., 2014).

4.7.1. Secretome analysis of activated macrophages 5. Development of therapeutics against AGEs


The most dramatically inducible proteins secreted by activated mac-
rophages are inflammatory molecules, such as cytokines and Developmental efforts have resulted in many agents that reduce the
chemokines. Among cytokines, surprisingly, various types of RAGE li- damage induced by AGEs. These agents can be summarized as AGEs in-
gands, such as AGEs (most abundantly in albumin), HMGB1 and S100 hibitors and breakers, angiotensin II receptor antagonists and angioten-
proteins, are secreted by activated macrophages. HMGB1 and S100 pro- sin converting enzyme inhibitors, antioxidants, natural substances, and
tein are damage-associated molecular pattern molecules (DAMPs) that anti-inflammatory molecules (Table 2).
are released by damaged and stressed cells. DAMPs, which are also
called alarmins or danger signals, are host-derived endogenous mole- 5.1. AGEs inhibitors & breakers
cules that can trigger immune response, and are recognized by DAMP
receptors, which include RAGE (Saïd-Sadier & Ojcius, 2012). AGEs inhibitors that prevent AGEs formation and AGEs cross-link
HMGB1 can be released from necrotic or damaged cells or by activat- breakers were identified as initial efforts. Aminoguanidine was the
ed immune cells, and regulates endothelial cell activation, cell migra- first AGEs inhibitor found to inhibit the formation of AGEs, and was
tion, inflammation, proliferation, and differentiation (Bertheloot & also reported to effectively reduce the pathologic effects of the nephrop-
Latz, 2016; Saïd-Sadier & Ojcius, 2012). In post-ischemic brains, athy and vascular complications associated with AGEs in diabetes
HMGB1 was found to be induced in and secreted by activated microglia (Bolton et al., 2004; Brownlee et al., 1986; Freedman et al., 1999). Sev-
(Kim et al., 2008; Kim et al., 2012; Shin et al., 2014). Interestingly, the eral AGEs cross-link breakers have also been reported to effectively re-
release and translocation of HMGB1 in macrophages were observed to duce AGEs products and proposed as possible therapeutics for the
be inhibited by various molecules that can reduce inflammation and treatment of diabetic vascular complications. Alagebrium and related
mortality in sepsis models (Cheng et al., 2015b; Elfeky et al., 2016; compounds including ALT-711, ALT-462, ALT-486, and ALT-946 appar-
Hagiwara et al., 2009; Kim et al., 2015; Ye et al., 2012; Zhang et al., ently break some existing AGEs cross-links (Krautwald et al., 2011).
2014; Zhou et al., 2014). HMGB1 secretion was also significantly in- ALT 711 was initially reported to effectively reduce the severity of
creased by macrophages in the presence of chronic inflammation dis- AGE-induced pathological lesions (Vasan et al., 2001). Another AGEs-
eases and in models of atherosclerosis (Kalinina et al., 2004), COPD breaker, TRC4186, was reported to inhibit AGEs formation or break
(Mortaz et al., 2015), rheumatoid arthritis (RA) (Zetterström et al., AGEs cross-links and to ameliorate diabetes-related cardiomyopathy
2008), and renal tubule-interstitial fibrosis (Tian et al., 2015). The and nephropathy (Joshi et al., 2009).
S100 protein family is composed of approximately 25 proteins (Gross
et al., 2014), and among them, S100A8 and S100 calcium binding pro- 5.2. Angiotensin II receptor antagonist and angiotensin converting enzyme
tein A9 (S100A9) are often found abundantly in macrophages (Farris inhibitor
et al., 2011; McCormick et al., 2005; Rammes et al., 1997; Xu & Geczy,
2000; Xu, Yen, & Geczy, 2001). These two proteins are also released by Telmisartan, an anti-hypertension drug and angiotensin II receptor
activated macrophages in patients with inflammatory muscle diseases, blocker, was found to decrease AGEs formation. For example, a clinical
and induced myoblast apoptosis via caspase 3 activation (Seeliger et study demonstrated that telmisartan decreased RAGE expression by ac-
al., 2003). tivating PPAR-γ in liver (Yoshida et al., 2006). In addition, pyridoxamine
has been reported as preventing the Amadori reaction leading to AGEs
4.7.2. Albumin and AGE-albumin synthesis in macrophages formation (Adrover et al., 2008; Thomas et al., 2005).
The liver is known as responsible for production of albumin in body,
however there are also reports of non-hepatic origins. These include 5.3. Antioxidants
pancreas, kidney (Nahon et al., 1988), skeletal muscle (Kobayashi &
Tashima, 1990; Müller & Heizmann, 1982), thyroid gland (De Vijlder Antioxidants have been developed to inhibit AGEs. Statins, which
et al., 1992). Activated macrophages are also known as alternate source have antioxidant activity, are commonly used to lower blood cholester-
which include microglial cells in AD (Byun et al., 2012a; 2012b), Alco- ol levels. The antioxidant effect of statins was found to inhibit reactive
holic brain damages (Byun et al., 2014) and PD (Bayarsaikhan et al., protein C release effectively from toxic AGEs, and thus, to prevent diabe-
2016). tes-associated vascular complications (Ajith et al., 2006, 2008; Takeuchi
Macrophage activation leads to the release RAGE ligands, including et al., 2010). Atorvastatin was introduced as means of decreasing serum
AGEs, HMGB1, amyloid beta (Aβ) and pro-inflammatory cytokines, AGEs levels in patients with type 2 diabetes in a soluble RAGE (sRAGE)
which at elevated levels damage host cells. These ligands are released and a splice variant of sRAGE dependent manner (Jinnouchi et al., 2006;
during inflammation or injury and this leads to the up-regulation of Tam et al., 2010). Other study supported the notion that atorvastatin re-
RAGE on host cells such as neurons (Borsi et al., 2012). Binding of stored reduced glutathione levels, inhibited RAGE up-regulation, and
AGEs to RAGE releases IL-β and TNF-α in AD (Fang et al., 2010), and down-regulated the expression of RAGE and MCP-1 (Feng et al.,
in PD regulates the expressions of pro-inflammatory cytokines and acti- 2011). Recently, a RAGE antagonist from the part of human serum albu-
vates of MAPK, NF-κB, and IFN-γ (Kim & Joh, 2006). Furthermore in AD, min was found to reduce the increased NF-κB signaling by anti-
50 K. Byun et al. / Pharmacology & Therapeutics 177 (2017) 44–55

inflammatory mechanism (Maltais et al., 2016). Ascorbic acid, alpha- peptide-1 receptor agonist exendin, and NF-κB inhibitor-pyrrolidine di-
lipoic acid, carnosine, and quercetin have been shown to inhibit AGEs thiocarbamate were also showed to provide strong in vitro protection
effectively (Abdul & Butterfield, 2007; Ashraf et al., 2015; Bierhaus et against AGEs-induced neuronal apoptosis by reducing the overexpres-
al., 1997; Guiotto et al., 2005; Zafar, 2012). Recently the glucagon-like sion of RAGE (Chen et al., 2016).

5.4. Natural substances


Table 2
Therapeutic agent for AGEs and RAGE related pathology in disease. Studies have shown that some natural substances, such as resvera-
trol and curcumin, can prevent AGEs-induced pathology (Mizutani et
Category Name Reference
al., 2000). Resveratrol inhibited the AGE-induced proliferation of colla-
AGEs inhibitor Aminoguanidine Brownlee, Vlassara,
gen in smooth muscle cells of blood vessels (Mizutani et al., 2000),
Kooney, Ulrich, and
Cerami (1986)
and curcumin blocked the effect of AGEs on the RAGE expression
Freedman et al. (Tang, 2014). Recently, new prenylflavonoids were reported to inhibit
(1999) the formations of N(6)-carboxymethyllysine (CML) and N(omega)-
Bolton et al. (2004) carboxymethylarginine (CMA) in the methanol extract of the aerial por-
AGEs cross link breaker Alagebrium and related Vasan, Foiles, and
tions of Epimedii Herba (Nakashima et al., 2016). Similarly, p-cymene, a
compounds (ALT-711, Founds (2001)
ALT-462, ALT-486, ALT- Krautwald et al. monoterpene was demonstrated to act as AGEs breakers for AGEs cross-
(2011) linking (Joglekar et al., 2014; Lee et al., 2014). Recently, sulphoraphene
TRC4186 Joshi et al. (2009) was demonstrated to inhibit inflammation in AGEs-exposed human
Angiotensin II receptor Telmisartan Yoshida et al. (2006)
umbilical vein endothelial cells (HUVECs) and AGEs-infused rat aorta
antagonist and angiotensin Pyridoxamine Thomas et al. (2005)
converting enzyme Adrover, Vilanova,
partly by suppressing RAGE expression (Matsui et al., 2016). Vitamin
inhibitor Frau, Muñoz, and D also reported as reducing the risk of CVDs. AGEs, such as CML, have
Donoso (2008) been implicated in diabetic vascular complications via oxidative
Antioxidant Statins Ajith, Harikumar, stress-mediated pathways (Salum et al., 2013). Gymnema also reported
Thasna, Sabu, and
to contribute to the anti-hyperglycemic effects (Sugihara et al., 2000),
Babitha (2006)
Ajith, Riji, and Anu and metformin has been reported to restore glycated high-density lipo-
(2008) protein (HDL)-mediated cholesterol efflux (Matsuki et al., 2009). Epi-
Takeuchi, Takino, and gallocatechin gallate (EGCG), a major component of green tea
Yamagishi (2010) polyphenols, showed protective effects against AGEs-induced dam-
Atorvastatin Jinnouchi et al. (2006)
Tam et al. (2010)
age in neuron cells (Lee & Lee, 2007), and S-allyl cysteine from
Feng et al. (2011) aged garlic has been reported to act as a potent antioxidant and in-
RAGE antagonist Maltais et al. (2016) hibit AGEs protein formation (Ahmad & Ahmed, 2006). Ramipril
Ascorbic acid Zafar (2012) has also been reported to be an efficient AGEs inhibitor as well
Alpha-lipoic acid Bierhaus et al. (1997)
(Gosse & Schumacher, 2014).
Abdul and Butterfield
(2007)
Carnosine Abdul and Butterfield 5.5. Anti-inflammatory molecules and AGEs inhibitors that act on activated
(2007) macrophages
Quercetin Ashraf et al. (2015)
Dhumale et al.
(2015)
Anti-inflammatory molecules have also been found to reduce
Exendin Chen et al. (2016) the toxic effects of AGEs. In particular, LR-90 had been shown to in-
Pyrrolidine Chen et al. (2016) hibit the NF-κB activation, which stimulates the gene expressions of
dithiocarbamate proinflammatory molecules in human monocytes (Figarola et al.,
Natural substance Resveratrol Mizutani, Ikeda, and
2007).
Yamori (2000)
Curcumin Tang (2014) Bisphosphonates were found to inhibit toxic AGEs-mediated inflam-
Prenylflavonoids Nakashima et al. mation and finally stopped ROS formation during the pathogeneses of
(2016) the vascular complications of diabetes (Takeuchi et al., 2010). Similarly,
p-Cymene Joglekar, Panaskar, another nitrogen-containing bisphosphonate, minodronate, inhibited
and Arvindekar
(2014)
signaling pathways initiated by the interaction between AGEs and
Chebulic acid of Terminalia Lee, Oh, Kim, and Lee RAGE in diabetic retinopathy (Yamagishi et al., 2006). It appears that in-
chebula, (2014) hibitors of AGEs with anti-inflammatory effects can deactivate macro-
Sulphoraphene Matsui, Nakamura, phages and reduce tissue damage as well.
Ojima, Nishino, and
Because most known therapeutic reagents used to treat diabetes
Yamagishi (2016)
Vitamin D Salum et al. (2013) target AGEs in blood, AGEs inhibition in activated macrophages lo-
Gymnema Sugihara et al. (2000) cated in tissues has been investigated as a novel targeting strategy.
Metformin Matsuki et al. (2009) The sRAGE-based masking of secreted AGEs has shown strong potent
Epigallocatechin Gallate Lee and Lee (2007) therapeutic effects in several diseases, such as AD, PD, alcoholic brain
S-allylcysteine Ahmad and Ahmed
(2006)
damage, and AMI (Bayarsaikhan et al., 2016; Byun et al., 2012a;
Ramipril Gosse and 2014) (Fig. 5). Since sRAGE is the extracellular part of RAGE, it can
Schumacher (2014) bind to AGEs, and thus, prevent their adverse effects. Targeting
Anti-inflammatory molecules LR-90 Figarola, AGEs in activated macrophage probably could be one of the most
Shanmugam,
ideal strategies for AGEs related diseases.
Natarajan, and
Rahbar (2007)
Bisphosphonates Takeuchi et al. (2010) 6. Conclusion and future perspectives
Minodronate Yamagishi,
Nakamura, Matsui, The role played by the AGEs-RAGE interaction in the aggravation
and Takeuchi (2006)
of the chronic complications of diabetes, CVDs, and degenerative
K. Byun et al. / Pharmacology & Therapeutics 177 (2017) 44–55 51

Fig. 5. A proposed model of AGE albumin from activated macrophage and its relevancy to cell death in various tissues.

diseases has been well established. The interaction between AGEs Acknowledgments
and RAGE on the plasma membrane triggers the downstream signal-
ing of inflammation, oxidative stress, and apoptosis in many cells, in- This work was supported by the Korean Health Technology R&D Pro-
cluding neurons, endothelial cells, lung cells and muscle cells. ject through the Korean Health Industry Development Institute (KHIDI),
Therapeutic agent development has focused mainly on suppressing funded by the Ministry of Health & Welfare, Republic of Korea (Lee, B.,
AGEs or RAGE formation or preventing the AGEs-RAGE interaction. grant no. HI13C2098), and by the National Research Foundation of Ko-
Although agents have been devised to prevent AGEs formation and rea (Byun, K., grant no. 2015R1A2A2A01005212).
activity, most are under in the early phase of clinical studies. Similar-
ly, studies on natural substances like resveratrol and curcumin have References
been described to inhibit RAGE-associated vascular damage and the
Abdelsalam, R. M., & Safar, M. M. (2015). Safar neuroprotective effects of vildagliptin in rat
long-term complications of diabetes. Furthermore, sRAGE and its rotenone Parkinson's disease model: Role of RAGE-NFκB and Nrf2-antioxidant signal-
variants can inhibit the interaction between AGEs and RAGE effec- ing pathways. Journal of Neurochemistry 133, 700–707.
tively and have also been suggested to be promising therapeutic Abdul, H. M., & Butterfield, D. A. (2007). Involvement of PI3K/PKG/ERK1/2 signaling path-
ways in cortical neurons to trigger protection by cotreatment of acetyl-L-carnitine
agents. and alpha-lipoic acid against HNE-mediated oxidative stress and neurotoxicity: Im-
Recently, it was found that activated macrophages are one of prima- plications for Alzheimer's disease. Free Radical Biology & Medicine 42, 371–384.
ry contributors to AGE-albumin accumulation in tissues, and suggested Abe, R., Shimizu, T., Sugawara, H., Watanabe, H., Nakamura, H., Choei, H., et al. (2004).
Regulation of human melanoma growth and metastasis by AGE-AGE receptor inter-
that AGE-albumin is a potentially useful therapeutic and diagnostic bio- actions. The Journal of Investigative Dermatology 122, 461–467.
marker with high sensitivity and resolution. In addition, labeling and vi- Adrover, M., Vilanova, B., Frau, J., Muñoz, F., & Donoso, J. (2008). The pyridoxamine action
sualizing accumulated AGE-albumin with proper antibodies may on Amadori compounds: A reexamination of its scavenging capacity and chelating ef-
fect. Bioorganic & Medicinal Chemistry 16, 5557–5569.
provide a near ideal theragnostic means to combat AGEs/RAGE associat- Ahmad, M. S., & Ahmed, N. (2006). Antiglycation properties of aged garlic extract: Possi-
ed diseases. ble role in prevention of diabetic complications. The Journal of Nutrition 136,
In conclusion, more studies are required to determine how to pre- 796S–799S.
Ahn, S. M., Byun, K., Cho, K., Kim, J. Y., Yoo, J. S., Kim, D., et al. (2008). Human microglial
vent, remove, and estimate the AGEs/RAGE interaction in relation to
cells synthesize albumin in brain. PloS One 3, e2829.
macrophage activation and to elucidate its pathophysiologic mecha- Ajith, T. A., Harikumar, K. B., Thasna, H., Sabu, M. C., & Babitha, N. V. (2006). Proapoptotic
nism in many intractable diseases. and antitumor activities of the HMG-CoA reductase inhibitor, lovastatin, against
Dalton's lymphoma ascites tumor in mice. Clinica Chimica Acta 366, 322–328.
Supplementary data to this article can be found online at http://dx.
Ajith, T. A., Riji, T., & Anu, V. (2008). In vitro anti-oxidant and DNA protective effects of the
doi.org/10.1016/j.pharmthera.2017.02.030. novel 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor rosuvastatin.
Clinical and Experimental Pharmacology & Physiology 35, 625–629.
Aleshin, A., Ananthakrishnan, R., Li, Q., Rosario, R., Lu, Y., Qu, W., et al. (2008). RAGE mod-
Conflict of interest statement
ulates myocardial injury consequent to LAD infarction via impact on JNK and STAT
signaling in a murine model. American Journal of Physiology. Heart and Circulatory
The authors declare that there are no conflicts of interest. Physiology 294, H1823–H1832.
52 K. Byun et al. / Pharmacology & Therapeutics 177 (2017) 44–55

Aliev, G., Palacios, H. H., Gasimov, E., Obrenovich, M. E., Morales, L., Leszek, J., et al. (2010). Donato, R. (2007). RAGE: A single receptor for several ligands and different cellular re-
Oxidative stress induced mitochondrial failure and vascular hypoperfusion as a key sponses: The case of certain S100 proteins. Current Molecular Medicine 7, 711–724.
initiator for the development of Alzheimer disease. Pharmaceuticals (Basel) 3, Dong, H., Li, J., Lv, Y., Zhou, Y., Wang, G., Hu, S., et al. (2013). Comparative analysis of the
158–187. alveolar macrophage proteome in ALI/ARDS patients between the exudative phase
Andrassy, M., Volz, H. C., Igwe, J. C., Funke, B., Eichberger, S. N., Kaya, Z., et al. (2008). High- and recovery phase. BMC Immunology 17, 14–25.
mobility group box-1 in ischemia-reperfusion injury of the heart. Circulation 117, Elfeky, M., Kaede, R., Okamatsu-Ogura, Y., & Kimura, K. (2016). Adiponectin inhibits LPS-
3216–3226. induced HMGB1 release through an AMP kinase and Heme oxygenase-1-dependent
Ansari, N. A., & Dash, D. (2013). Amadori glycated proteins: Role in production of autoan- pathway in RAW 264 macrophage cells. Mediators of Inflammation 2016, 5701959.
tibodies in diabetes mellitus and effect of inhibitors on non-enzymatic glycation. Fang, F., Lue, L. F., Yan, S., Xu, H., Luddy, J. S., Chen, D., et al. (2010). RAGE-dependent sig-
Aging and Disease 4, 50–56. naling in microglia contributes to neuroinflammation, Aβ accumulation, and im-
Arsov, S., Graaff, R., van Oeveren, W., Stegmayr, B., Sikole, A., Rakhorst, G., et al. (2014). paired learning/memory in a mouse model of Alzheimer's disease. The FASEB
Advanced glycation end-products and skin autofluorescence in end-stage renal dis- Journal 24, 1043–1055.
ease: A review. Clinical Chemistry and Laboratory Medicine 52, 11–20. Farris, S. D., Hu, J. H., Krishnan, R., Emery, I., Chu, T., Du, L., et al. (2011). Mechanisms of
Ashraf, J. M., Shahab, U., Tabrez, S., Lee, E. J., Choi, I., & Ahmad, S. (2015). Quercetin as a urokinase plasminogen activator (uPA)-mediated atherosclerosis: Role of the uPA re-
finer substitute to aminoguanidine in the inhibition of glycation products. ceptor and S100A8/A9 proteins. The Journal of Biological Chemistry 286, 22665–22677.
International Journal of Biological Macromolecules 77, 188–192. Feng, B., Xu, L., Wang, H., Yan, X., Xue, J., Liu, F., et al. (2011). Atorvastatin exerts its anti-
Bayarsaikhan, E., Bayarsaikhan, D., Lee, J., Son, M., Oh, S., Moon, J., et al. (2016). Microglial atherosclerotic effects by targeting the receptor for advanced glycation end products.
AGE-albumin is critical for neuronal death in Parkinson's disease: A possible implica- Biochimica et Biophysica Acta 1812, 1130–1137.
tion for theranostics. International Journal of Nanomedicine 23(Spec Iss), 281–292. Figarola, J. L., Shanmugam, N., Natarajan, R., & Rahbar, S. (2007). Anti-inflammatory ef-
Bertheloot, D., & Latz, E. (2016). HMGB1, IL-1α, IL-33 and S100 proteins: Dual-function fects of the advanced glycation end product inhibitor LR-90 in human monocytes.
alarmins. Cellular & Molecular Immunology 13, 1–22. Diabetes 56, 647–655.
Bierhaus, A., Chevion, S., Chevion, M., Hofmann, M., Quehenberger, P., Illmer, T., et al. Freeburn, R. W., Armstrong, L., & Millar, A. B. (2005). Cultured alveolar macrophages from
(1997). Advanced glycation end product-induced activation of NF-κB is suppressed patients with idiopathic pulmonary fibrosis (IPF) show dysregulation of lipopolysac-
by α-lipoic acid in cultured endothelial cells. Diabetes 46, 1481–1490. charide-induced tumor necrosis factor-alpha (TNF-alpha) and interleukin-10 (IL-10)
Bierhaus, A., Hofmann, M. A., Ziegler, R., & Nawroth, P. P. (1998). AGEs and their interac- inductions. European Cytokine Network 16, 5–16.
tion with AGE-receptors in vascular disease and diabetes mellitus. I. The AGE concept. Freedman, B. I., Wuerth, J. P., Cartwright, K., Bain, R. P., Dippe, S., Hershon, K., et al. (1999).
Cardiovascular Research 37, 586–600. Design and baseline characteristics for the aminoguanidine clinical trial in overt type
Blanco, F. J., López-Armada, M. J., & Maneiro, E. (2004). Mitochondrial dysfunction in os- 2 diabetic nephropathy (ACTION II). Controlled Clinical Trials 20, 493–510.
teoarthritis. Mitochondrion 4, 715–728. Freiwald, A., Weidner, C., Witzke, A., Huang, S. Y., Meierhofer, D., & Sauer, S. (2013). Com-
Bolton, W. K., Cattran, D. C., Williams, M. E., Adler, S. G., Appel, G. B., Cartwright, K., et al. prehensive proteomic data sets for studying adipocyte-macrophage cell-cell commu-
(2004). Randomized trial of an inhibitor of formation of advanced glycation end nication. Proteomics 13, 3424–3428.
products in diabetic nephropathy. American Journal of Nephrology 24, 32–40. Ge, J., Jia, Q., Liang, C., Luo, Y., Huang, D., Sun, A., et al. (2005). Advanced glycosylation end
Borsi, V., Cerofolini, L., Fragai, M., & Luchinat, C. (2012). NMR characterization of the C-ter- products might promote atherosclerosis through inducing the immune maturation of
minal tail of full-length RAGE in a membrane mimicking environment. Journal of dendritic cells. Arteriosclerosis, Thrombosis, and Vascular Biology 25, 2157–2163.
Biomolecular NMR 54, 285–290. Gensel, J. C., & Zhang, B. (2015). Macrophage activation and its role in repair and pathol-
Bortolotto, V., & Grilli, M. (2016). Every cloud has a silver lining: Proneurogenic effects of ogy after spinal cord injury. Brain Research 1619, 1–11.
Aβ oligomers and HMGB-1 via activation of the RAGE-NF-κB axis. CNS & Neurological Gkogkolou, P., & Böhm, M. (2012). Advanced glycation end products: Key players in skin
Disorders Drug Targets 15, 1–14. aging? Dermatoendocrinology 4, 259–270.
Brass, L. F., & Molino, M. (1997). Protease-activated G protein-coupled receptors on Goldin, A., Beckman, J. A., Schmidt, A. M., & Creager, M. A. (2006). Advanced glycation end
human platelets and endothelial cells. Thrombosis and Haemostasis 78, 234–241. products: Sparking the development of diabetic vascular injury. Circulation 114,
Brownlee, M., Vlassara, H., Kooney, A., Ulrich, P., & Cerami, A. (1986). Aminoguanidine 597–605.
prevents diabetes-induced arterial wall protein cross-linking. Science 232, Gosse, P., & Schumacher, H. (2014). Effect of telmisartan vs. ramipril on ‘dipping’ status
1629–1632. and blood pressure variability: Pooled analysis of the PRISMA studies. Hypertension
Burillo, E., Recalde, D., Jarauta, E., Fiddyment, S., Garcia-Otin, A. L., Mateo-Gallego, R., et al. Research 37, 151–157.
(2009). Proteomic study of macrophages exposed to oxLDL identifies a CAPG poly- Gross, S. R., Sin, C. G., Barraclough, R., & Rudland, P. S. (2014). Joining S100 proteins and
morphism associated with carotid atherosclerosis. Atherosclerosis 207, 32–37. migration: For better or for worse, in sickness and in health. Cellular and Molecular
Byun, K., Bayarsaikhan, E., Kim, D., Kim, C. Y., Mook-Jung, I., Paek, S. H., et al. (2012a). In- Life Sciences 71, 1551–1579.
duction of neuronal death by microglial AGE-albumin: Implications for Alzheimer's Gruys, E., Toussaint, M. J., Niewold, T. A., & Koopmans, S. J. (2005). Acute phase reaction
disease. PloS One 7, e37917. and acute phase proteins. Journal of Zhejiang University. Science. B 6, 1045–1056.
Byun, K., Bayarsaikhan, E., Kim, D., Son, M., Hong, J., Jeong, G. B., et al. (2012b). Activated Guiotto, A., Calderan, A., Ruzza, P., & Borin, G. (2005). Carnosine and carnosine-related an-
microglial cells synthesize and secrete AGE-albumin. Anatomy and Cell Biology 45, tioxidants: A review. Current Medicinal Chemistry 12, 2293–2315.
47–52. Hagiwara, S., Iwasaka, H., Matumoto, S., Hidaka, S., & Noguchi, T. (2009). Effects of an an-
Byun, K., Bayarsaikhan, D., Bayarsaikhan, E., Son, M., Oh, S., Lee, J., et al. (2014). Microglial giotensin-converting enzyme inhibitor on the inflammatory response in in vivo and
AGE-albumin is critical in promoting alcohol-induced neurodegeneration in rats and in vitro models. Critical Care Medicine 37, 626–633.
humans. PloS One 9, e104699. He, M., Kubo, H., Ishizawa, K., Hegab, A. E., Yamamoto, Y., Yamamoto, H., et al. (2007). The
Chavakis, T., Bierhaus, A., & Nawroth, P. P. (2004). RAGE (receptor for advanced glycation role of the receptor for advanced glycation end-products in lung fibrosis. American
end products): A central player in the inflammatory response. Microbes and Infection Journal of Physiology. Lung Cellular and Molecular Physiology 293, L1427–L1436.
6, 1219–1225. He, M., Kubo, H., Morimoto, K., Fujino, N., Suzuki, T., Takahasi, T., et al. (2011). Receptor for
Chen, S., Yin, L., Xu, Z., An, F. M., Liu, A. R., Wang, Y., et al. (2016). Inhibiting receptor for advanced glycation end products binds to phosphatidylserine and assists in the clear-
advanced glycation end product (AGE) and oxidative stress involved in the protective ance of apoptotic cells. EMBO Reports 12, 358–364.
effect mediated by glucagon-like peptide-1 receptor on AGE induced neuronal apo- Heneka, M. T., O'Banion, M. K., Terwel, D., & Kummer, M. P. (2010). Neuroinflammatory
ptosis. Neuroscience Letters 612, 193–198. processes in Alzheimer's disease. Journal of Neural Transmission (Vienna) 117,
Cheng, M., Liu, H., Zhang, D., Liu, Y., Wang, C., Liu, F., & Chen, J. (2015a). HMGB1 enhances 919–947.
the AGE-induced expression of CTGF and TGF-β via RAGE-dependent signaling in Hernanz, R., Briones, A. M., Alonso, M. J., Vila, E., & Salaices, M. (2004). Hypertension alters
renal tubular epithelial cells. American Journal of Nephrology 41, 257–266. role of iNOS, COX-2, and oxidative stress in bradykinin relaxation impairment after
Cheng, Y., Wang, H., Mao, M., Liang, C., Zhang, Y., Yang, D., et al. (2015b). Escin increases LPS in rat cerebral arteries. American Journal of Physiology. Heart and Circulatory
the survival rate of LPS-induced septic mice through inhibition of HMGB1 release Physiology 287, H225–H234.
from macrophages. Cellular Physiology and Biochemistry 36, 1577–1586. Herzog, C., Lorenz, A., Gillmann, H. J., Chowdhury, A., Larmann, J., Harendza, T., et al.
Crews, F. T., & Vetreno, R. P. (2014). Neuroimmune basis of alcoholic brain damage. (2014). Thrombomodulin's lectin-like domain reduces myocardial damage by
International Review of Neurobiology 118, 315–357. interfering with HMGB1-mediated TLR2 signalling. Cardiovascular Research 101,
Crews, F. T., & Vetreno, R. P. (2016). Mechanisms of neuroimmune gene induction in al- 400–410.
coholism. Psychopharmacology 233, 1543–1557. Hirohashi, N., & Morrison, D. C. (1996). Low-dose lipopolysaccharide (LPS) pretreatment
De Vijlder, J. J., Veenboer, G. J., & van Dijk, J. E. (1992). Thyroid albumin originates from of mouse macrophages modulates LPS-dependent interleukin-6 production in vitro.
blood. Endocrinology 131, 578–584. Infection and Immunity 64, 1011–1015.
Deng, Y., Yang, Z., Gao, Y., Xu, H., Zheng, B., Jiang, M., et al. (2013). Toll-like receptor 4 me- Hofmann, M. A., Drury, S., Fu, C., Qu, W., Taguchi, A., Lu, Y., et al. (1999). RAGE mediates a
diates acute lung injury induced by high mobility group box-1. PloS One 8, e64375. novel proinflammatory axis: A central cell surface receptor for S100/calgranulin poly-
Dhumale, S. S., Waghela, B. N., & Pathak, C. (2015). Quercetin protects necrotic insult and peptides. Cell 97, 889–901.
promotes apoptosis by attenuating the expression of RAGE and its ligand HMGB1 in Huttunen, H. J., Fages, C., & Rauvala, H. (1999). Receptor for advanced glycation end prod-
human breast adenocarcinoma cells. IUBMB Life 67, 361–373. ucts (RAGE)-mediated neurite outgrowth and activation of NF-κB require the cyto-
Di Filippo, C., Cuzzocrea, S., Rossi, F., Marfella, R., & D'Amico, M. (2006). Oxidative stress as plasmic domain of the receptor but different downstream signaling pathways. The
the leading cause of acute myocardial infarction in diabetics. Cardiovascular Drug Journal of Biological Chemistry 274, 19919–19924.
Reviews 24, 77–87. Jain, S., Gautam, V., & Naseem, S. (2011). Acute-phase proteins: As diagnostic tool. Journal
Dias, V., Junn, E., & Mouradian, M. M. (2013). The role of oxidative stress in Parkinson's of Pharmacy & Bioallied Sciences 3, 118–127.
disease. Journal of Parkinson's Disease 3, 461–491. Jinnouchi, Y., Yamagishi, S., Takeuchi, M., Ishida, S., Jinnouchi, Y., Jinnouchi, J., et al. (2006).
Donato, R. (1999). Functional roles of S100 proteins, calcium-binding proteins of the EF- Atorvastatin decreases serum levels of advanced glycation end products (AGEs) in
hand type. Biochimica et Biophysica Acta 1450, 191–231. patients with type 2 diabetes. Clinical and Experimental Medicine 6, 191–193.
K. Byun et al. / Pharmacology & Therapeutics 177 (2017) 44–55 53

Jirillo, E., Caccavo, D., Magrone, T., Piccigallo, E., Amati, L., Lembo, A., et al. (2002). The role Limongi, D., & Baldelli, S. (2016). Redox imbalance and viral infections in neurodegener-
of the liver in the response to LPS: Experimental and clinical findings. Journal of ative diseases. Oxidative Medicine and Cellular Longevity 2016, 6547248.
Endotoxin Research 8, 319–327. Luevano-Contreras, C., & Chapman-Novakofski, K. (2010). Dietary advanced glycation end
Joglekar, M. M., Panaskar, S. N., & Arvindekar, A. U. (2014). Inhibition of advanced products and aging. Nutrients 2, 1247–1265.
glycation end product formation by cymene–A common food constituent. Journal of Lv, C., Wang, L., Liu, X., Cong, X., Yan, S. S., Wang, Y., et al. (2014). Geniposide attenuates
Functional Foods 6, 107–115. oligomeric Aβ(1-42)-induced inflammatory response by targeting RAGE-dependent
Jono, T., Kimura, T., Takamatsu, J., Nagai, R., Miyazaki, K., Yuzuriha, T., et al. (2002). Accu- signaling in BV2 cells. Current Alzheimer Research 11, 430–440.
mulation of imidazolone, pentosidine and N(epsilon)-(carboxymethyl) lysine in hip- Ma, Y., Bao, J., Zhao, X., Shen, H., Lv, J., Ma, S., et al. (2013). Activated cyclin-dependent ki-
pocampal CA4 pyramidal neurons of aged human brain. Pathology International 52, nase 5 promotes microglial phagocytosis of fibrillar β-amyloid by up-regulating lipo-
563–571. protein lipase expression. Molecular & Cellular Proteomics 12, 2833–2844.
Joshi, D., Gupta, R., Dubey, A., Shiwalkar, A., Pathak, P., Gupta, R. C., et al. (2009). Macdonald, J., Galley, H. F., & Webster, N. R. (2003). Oxidative stress and gene expression
TRC4186, a novel AGE-breaker, improves diabetic cardiomyopathy and nephrop- in sepsis. British Journal of Anaesthesia 90, 221–232.
athy in Ob-ZSF1 model of type 2 diabetes. Journal of Cardiovascular Pharmacology Mahali, S., Raviprakash, N., Raghavendra, P. B., & Manna, S. K. (2011). Advanced glycation
54, 72–81. end products (AGEs) induce apoptosis via a novel pathway: Involvement of Ca2+
Kalinina, N., Agrotis, A., Antropova, Y., DiVitto, G., Kanellakis, P., Kostolias, G., et al. (2004). mediated by interleukin-8 protein. The Journal of Biological Chemistry 286 (34903-
Increased expression of the DNA-binding cytokine HMGB1 in human atherosclerotic 34013).
lesions: Role of activated macrophages and cytokines. Arteriosclerosis, Thrombosis, and Maltais, J. S., Simard, E., Froehlich, U., Denault, J. B., Gendron, L., & Grandbois, M. (2016).
Vascular Biology 24, 2320–2325. iRAGE as a novel carboxymethylated peptide that prevents advanced glycation end
Kang, J. H., Kim, H. T., Choi, M. S., Lee, W. H., Huh, T. L., Park, Y. B., et al. (2006). Proteome product-induced apoptosis and endoplasmic reticulum stress in vascular smooth
analysis of human monocytic THP-1 cells primed with oxidized low-density lipopro- muscle cells. Pharmacological Research 104, 176–185.
teins. Proteomics 6, 1261–1273. Matsui, T., Nakamura, N., Ojima, A., Nishino, Y., & Yamagishi, S. I. (2016). Sulforaphane re-
Kao, Y. H., Lin, Y. C., Tsai, M. S., Sun, C. K., Yuan, S. S., Chang, C. Y., et al. (2014). Involvement duces advanced glycation end products (AGEs)-induced inflammation in endothelial
of the nuclear high mobility group B1 peptides released from injured hepatocytes in cells and rat aorta. Nutrition, Metabolism, and Cardiovascular Diseases 26, 797–807.
murine hepatic fibrogenesis. Biochimica et Biophysica Acta 1842, 1720–1732. Matsuki, K., Tamasawa, N., Yamashita, M., Tanabe, J., Murakami, H., Matsui, J., et al. (2009).
Karumanchi, D. K., Karunaratne, N., Lurio, L., Dillon, J. P., & Gaillard, E. R. (2015). Non-en- Metformin restores impaired HDL-mediated cholesterol efflux due to glycation.
zymatic glycation of α-crystallin as an in vitro model for aging, diabetes and degen- Atherosclerosis 206, 434–438.
erative diseases. Amino Acids 47, 2601–2608. Mayer, B., John, M., & Böhme, E. (1990). Purification of a Ca2+/calmodulin-dependent ni-
Kikuchi, K., Uchikado, H., Miura, N., Morimoto, Y., Ito, T., Tancharoen, S., et al. (2011). tric oxide synthase from porcine cerebellum. FEBS Letters 277, 215–219.
HMGB1 as a therapeutic target in spinal cord injury: A hypothesis for novel therapy McCormick, M. M., Rahimi, F., Bobryshev, Y. V., Gaus, K., Zreiqat, H., Cai, H., et al. (2005).
development. Experimental and Therapeutic Medicine 2, 767–770. S100A8 and S100A9 in human arterial wall. Implications for atherogenesis. Journal
Kim, Y. S., & Joh, T. H. (2006). Microglia, major player in the brain inflammation: Their roles in of Biological Chemistry 280, 41521–41529.
the pathogenesis of Parkinson's disease. Experimental & Molecular Medicine 38, 333–347. Miettinen, J. J., Matikainen, S., & Nyman, T. A. (2012). Global secretome characterization of
Kim, J. M., Han, H. J., Hur, Y. H., Quan, H., Kwak, S. H., Choi, J. I., et al. (2015). Stearoyl herpes simplex virus 1-infected human primary macrophages. Journal of Virology 86,
lysophosphatidylcholine prevents lipopolysaccharide-induced extracellular release 12770–12778.
of high mobility group box-1 through AMP-activated protein kinase activation. Miles, P. R., Bowman, L., Rao, K. M., Baatz, J. E., & Huffman, L. (1999). Pulmonary surfactant
International Immunopharmacology 28, 540–545. inhibits LPS-induced nitric oxide production by alveolar macrophages. The American
Kim, S. W., Jin, Y., Shin, J. H., Kim, I. D., Lee, H. K., Park, S., et al. (2012). Glycyrrhizic acid Journal of Physiology 276, L186–L196.
affords robust neuroprotection in the postischemic brain via anti-inflammatory effect Miyazaki, A., Nakayama, H., & Horiuchi, S. (2002). Scavenger receptors that recognize ad-
by inhibiting HMGB1 phosphorylation and secretion. Neurobiology of Disease 46, vanced glycation end products. Trends in Cardiovascular Medicine 12, 258–262.
147–156. Mizutani, K., Ikeda, K., & Yamori, Y. (2000). Resveratrol inhibits AGEs-induced prolifera-
Kim, J. B., Lim, C. M., Yu, Y. M., & Lee, J. K. (2008). Induction and subcellular localization of tion and collagen synthesis activity in vascular smooth muscle cells from stroke-
high-mobility group box-1 (HMGB1) in the postischemic rat brain. Journal of prone spontaneously hypertensive rats. Biochemical and Biophysical Research
Neuroscience Research 86, 1125–1131. Communications 274, 61–67.
Kobayashi, R., & Tashima, Y. (1990). Purification, biological properties and partial se- Mortaz, E., Adcock, I. M., Ricciardolo, F. L., Varahram, M., Jamaati, H., Velayati, A. A., et al.
quence analysis of 67-kDa calcimedin and its 34-kDa fragment from chicken gizzard. (2015). Anti-inflammatory effects of Lactobacillus rahmnosus and Bifidobacterium
European Journal of Biochemistry 188, 447–453. breve on cigarette smoke activated human macrophages. PloS One 10, e0136455.
Kobayashi, R., Deavers, M., Patenia, R., Rice-Stitt, T., Halbe, J., Gallardo, S., et al. (2009). 14- Müller, G., & Heizmann, C. W. (1982). Albumin in chicken skeletal muscle. European
3-3 zeta protein secreted by tumor associated monocytes/macrophages from ascites Journal of Biochemistry 123, 577–582.
of epithelial ovarian cancer patients. Cancer Immunology, Immunotherapy 58, 247–258. Nahon, J. L., Tratner, I., Poliard, A., Presse, F., Poiret, M., Gal, A., et al. (1988). Albumin and
Kokkola, R., Sundberg, E., Ulfgren, A. K., Palmblad, K., Li, J., Wang, H., et al. (2002). High alpha-fetoprotein gene expression in various nonhepatic rat tissues. The Journal of
mobility group box chromosomal protein 1: A novel proinflammatory mediator in Biological Chemistry 263, 11436–11442.
synovitis. Arthritis and Rheumatism 46, 2598–2603. Nakamura, T., Sato, E., Fujiwara, N., Kawagoe, Y., Ueda, Y., Suzuki, T., et al. (2009). Positive
Kraft, A. D., & Harry, G. J. (2011). Features of microglia and neuroinflammation relevant to association of serum levels of advanced glycation end products and high mobility
environmental exposure and neurotoxicity. International Journal of Environmental group box-1 with asymmetric dimethylarginine in nondiabetic chronic kidney dis-
Research and Public Health 8, 2980–3018. ease patients. Metabolism 58, 1624–1628.
Kratz, M., Coats, B. R., Hisert, K. B., Hagman, D., Mutskov, V., Peris, E., et al. (2014). Meta- Nakashima, K., Miyashita, H., Yoshimitsu, H., Fujiwara, Y., Nagai, R., & Ikeda, T. (2016).
bolic dysfunction drives a mechanistically distinct proinflammatory phenotype in ad- Two new prenylflavonoids from Epimedii herba and their inhibitory effects on ad-
ipose tissue macrophages. Cell Metabolism 20, 614–625. vanced glycation end-products. Journal of Natural Medicines 70, 290–295.
Krautwald, M., Leech, D., Horne, S., Steele, M. L., Forbes, J., Rahmadi, A., et al. (2011). The Nakashima, M., Sakai, T., Hiraiwa, H., Hamada, T., Omachi, T., Ono, Y., et al. (2012). Role of
advanced glycation end product-lowering agent ALT-711 is a low-affinity inhibitor of S100A12 in the pathogenesis of osteoarthritis. Biochemical and Biophysical Research
thiamine diphosphokinase. Rejuvenation Research 14, 383–391. Communications 422, 508–514.
Kuhla, A., Trieglaff, C., & Vollmar, B. (2011). Role of age and uncoupling protein-2 in oxi- Nienhuis, H. L., Westra, J., Smit, A. J., Limburg, P. C., Kallenberg, C. G., & Bijl, M. (2009). AGE
dative stress, RAGE/AGE interaction and inflammatory liver injury. Experimental and their receptor RAGE in systemic autoimmune diseases: An inflammation propa-
Gerontology 46, 868–876. gating factor contributing to accelerated atherosclerosis. Autoimmunity 42, 302–304.
Kurien, B. T., & Scofield, R. H. (2008). Autoimmunity and oxidatively modified Nystedt, S., Ramakrishnan, V., & Sundelin, J. (1996). The proteinase-activated receptor 2 is
autoantigens. Autoimmunity Reviews 7, 567–573. induced by inflammatory mediators in human endothelial cells comparison with the
Lane, R. K., Hilsabeck, T., & Rea, S. L. (2015). The role of mitochondrial dysfunction in age- thrombin receptor. The Journal of Biological Chemistry 271, 14910–14915.
related diseases. Biochimica et Biophysica Acta 1847, 1387–1400. Origlia, N., Bonadonna, C., Rosellini, A., Leznik, E., Arancio, O., Yan, S. S., et al. (2010).
Lee, S. -J., & Lee, K. -W. (2007). Protective effect of (-)-epigallocatechin gallate against ad- Microglial receptor for advanced glycation end product-dependent signal pathway
vanced glycation endproducts-induced injury in neuronal cells. Biol Pharm Bull 30(8), drives β-amyloid-induced synaptic depression and long-term depression impair-
1369–1373. ment in entorhinal cortex. The Journal of Neuroscience 30, 11414–11425.
Lee, J. Y., Oh, J. G., Kim, J. S., & Lee, K. W. (2014). Effects of chebulic acid on advanced Ott, C., Jacobs, K., Haucke, E., Navarrete Santos, A., Grune, T., & Simm, A. (2014). Role of
glycation endproducts-induced collagen cross-links. Biological & Pharmaceutical advanced glycation end products in cellular signaling. Redox Biology 2, 411–429.
Bulletin 37, 1162–1167. Rahman, I. (2005). The role of oxidative stress in the pathogenesis of COPD. Treatments in
Legler, D. F., Bruckner, M., Uetz-von Allmen, E., & Krause, P. (2010). Prostaglandin E2 at Respiratory Medicine 4, 175–200.
new glance: Novel insights in functional diversity offer therapeutic chances. The Ramasamy, R., Yan, S. F., & Schmidt, A. M. (2012). Advanced glycation endproducts: From
International Journal of Biochemistry & Cell Biology 42, 198–201. precursors to RAGE: Round and round we go. Amino Acids 42, 1151–1161.
Li, Y., Takemura, G., Kosai, K. I., Takahashi, T., Okada, H., Miyata, S., et al. (2004). Critical Rammes, A., Roth, J., Goebeler, M., Klempt, M., Hartmann, M., & Sorg, C. (1997).
roles for the Fas/Fas ligand system in postinfarction ventricular remodeling and Myeloidrelated protein (MRP) 8 and MRP14, calcium-binding proteins of the S100
heart failure. Circulation Research 95, 627–636. family, are secreted by activated monocytes via a novel, tubulin-dependent pathway.
Liesz, A., Dalpke, A., Mracsko, E., Antoine, D. J., Roth, S., Zhou, W., et al. (2015). DAMP sig- The Journal of Biological Chemistry 272, 9496–9502.
naling is a key pathway inducing immune modulation after brain injury. The Journal Reynolds, A. D., Glanzer, J. G., Kadiu, I., Ricardo-Dukelow, M., Chaudhuri, A., Ciborowski, P.,
of Neuroscience 35, 583–598. et al. (2008a). Nitrated alpha-synuclein-activated microglial profiling for Parkinson's
Lietzén, N., Ohman, T., Rintahaka, J., Julkunen, I., Aittokallio, T., Matikainen, S., et al. (2011). disease. Journal of Neurochemistry 104, 1504–1525.
Quantitative subcellular proteome and secretome profiling of influenza A virus-in- Reynolds, P. R., Kasteler, S. D., Cosio, M. G., Sturrock, A., Huecksteadt, T., & Hoidal, J. R.
fected human primary macrophages. PLoS Pathogens 7, e1001340. (2008b). RAGE: Developmental expression and positive feedback regulation by Egr-
54 K. Byun et al. / Pharmacology & Therapeutics 177 (2017) 44–55

1 during cigarette smoke exposure in pulmonary epithelial cells. American Journal of Tessier, F., Obrenovich, M., & Monnier, V. M. (1999). Structure and mechanism of forma-
Physiology. Lung Cellular and Molecular Physiology 294, L1094–L1101. tion of human lens fluorophore LM-1. Relationship to vesperlysine A and the ad-
Ricciotti, E., & FitzGerald, G. A. (2011). Prostaglandins and inflammation. Arteriosclerosis, vanced Maillard reaction in aging, diabetes, and cataractogenesis. The Journal of
Thrombosis, and Vascular Biology 31, 986–1000. Biological Chemistry 274, 20796–20804.
Ritchie, R. F., Palomaki, G. E., Neveux, L. M., Navolotskaia, O., Ledue, T. B., & Craig, W. Y. Thomas, M. C., Tikellis, C., Burns, W. M., Bialkowski, K., Cao, Z., Coughlan, M. T., et al.
(1999). Reference distributions for the negative acute-phase serum proteins, albu- (2005). Interactions between renin angiotensin system and advanced glycation in
min, transferrin and transthyretin: A practical, simple and clinically relevant ap- the kidney. Journal of the American Society of Nephrology 16, 2976–2984.
proach in a large cohort. Journal of Clinical Laboratory Analysis 13, 273–279. Tian, S., Zhang, L., Tang, J., Guo, X., Dong, K., & Chen, S. Y. (2015). HMGB1 exacerbates renal
Rolo, A. P., & Palmeira, C. M. (2006). Diabetes and mitochondrial function: Role of hy- tubulointerstitial fibrosis through facilitating M1 macrophage phenotype at the early
perglycemia and oxidative stress. Toxicology and Applied Pharmacology 212, stage of obstructive injury. American Journal of Physiology. Renal Physiology 308,
167–178. F69–F75.
Russo, I., & Frangogiannis, N. G. (2016). Diabetes-associated cardiac fibrosis: Cellular ef- Tsoporis, J. N., Izhar, S., Leong-Poi, H., Desjardins, J. F., Huttunen, H. J., & Parker, T. G.
fectors, molecular mechanisms and therapeutic opportunities. Journal of Molecular (2010). S100B interaction with the receptor for advanced glycation end products
and Cellular Cardiology 90, 84–93. (RAGE): A novel receptor-mediated mechanism for myocyte apoptosis
Ryan, B. J., Nissim, A., & Winyard, P. G. (2014). Oxidative post-translational modifications postinfarction. Circulation Research 106, 93–101.
and their involvement in the pathogenesis of autoimmune diseases. Redox Biology 28, van Lent, P. L., Blom, A. B., Schelbergen, R. F., Slöetjes, A., Lafeber, F. P., Lems, W. F., et al.
715–724. (2012). Active involvement of alarmins S100A8 and S100A9 in the regulation of sy-
Saïd-Sadier, N., & Ojcius, D. M. (2012). Alarmins, inflammasomes and immunity. novial activation and joint destruction during mouse and human osteoarthritis.
Biomedical Journal 35, 437–449. Arthritis and Rheumatism 64, 1466–1476.
Salum, E., Kals, J., Kampus, P., Salum, T., Zilmer, K., Aunapuu, M., et al. (2013). Vitamin D Varol, C., Mildner, A., & Jung, S. (2015). Macrophages: Development and tissue specializa-
reduces deposition of advanced glycation end-products in the aortic wall and system- tion. Annual Review of Immunology 33, 643–675.
ic oxidative stress in diabetic rats. Diabetes Research and Clinical Practice 100, Vasan, S., Foiles, P. G., & Founds, H. W. (2001). Therapeutic potential of AGE inhibitors and
243–249. breakers of AGE protein cross-links. Expert Opinion on Investigational Drugs 10,
Sathe, K., Maetzler, W., Lang, J. D., Mounsey, R. B., Fleckenstein, C., Martin, H. L., et al. 1977–1987.
(2012). S100B is increased in Parkinson's disease and ablation protects against Wallace, J. L. (2005). Nitric oxide as a regulator of inflammatory processes. Memórias do
MPTP-induced toxicity through the RAGE and TNF-α pathway. Brain 135, Instituto Oswaldo Cruz 100, 5–9.
3336–3347. Wang, C. Y., Xie, J. W., Xu, Y., Wang, T., Cai, J. H., Wang, X., et al. (2013). Trientine reduces
Schmidt, A. M., Du Yan, S., Yan, S. F., & Stern, D. M. (2001). The multiligand receptor RAGE BACE1 activity and mitigates amyloidosis via the AGE/RAGE/NF-κB pathway in a
as a progression factor amplifying immune and inflammatory responses. The Journal transgenic mouse model of Alzheimer's disease. Antioxidants & Redox Signaling 19,
of Clinical Investigation 108, 949–955. 2024–2039.
Seeliger, S., Vogl, T., Engels, I. H., Schröder, J. M., Sorg, C., Sunderkötter, C., et al. (2003). Ex- Wang, X., Yu, S., Hu, J. P., Wang, C. Y., Wang, Y., Liu, H. X., et al. (2014). Streptozotocin-in-
pression of calcium-binding proteins MRP8 and MRP14 in inflammatory muscle dis- duced diabetes increases amyloid plaque deposition in AD transgenic mice through
eases. The American Journal of Pathology 163, 947–956. modulating AGEs/RAGE/NF-κB pathway. The International Journal of Neuroscience
Shin, J. H., Lee, H. K., Lee, H. B., Jin, Y., & Lee, J. K. (2014). Ethyl pyruvate inhibits HMGB1 124, 601–608.
phosphorylation and secretion in activated microglia and in the postischemic brain. Wang, L., Zhang, X., Liu, L., Yang, R., Cui, L., & Li, M. (2010). Atorvastatin protects rat brains
Neuroscience Letters 558, 159–163. against permanent focal ischemia and downregulates HMGB1, HMGB1 receptors
Simm, A., Wagner, J., Gursinsky, T., Nass, N., Friedrich, I., Schinzel, R., et al. (2007). Ad- (RAGE and TLR4), NF-kappaB expression. Neuroscience Letters 471, 152–156.
vanced glycation endproducts: A biomarker for age as an outcome predictor after car- Ward, M. S., Fortheringham, A. K., Cooper, M. E., & Forbes, J. M. (2013). Targeting ad-
diac surgery? Experimental Gerontology 42, 668–675. vanced glycation endproducts and mitochondrial dysfunction in cardiovascular dis-
Singh, R., Barden, A., Mori, T., & Beilin, L. (2001). Advanced glycation end-products: A re- ease. Current Opinion in Pharmacology 13, 654–661.
view. Diabetologia 44, 129–146. Xie, Z., Smith, C. J., & Van Eldik, L. J. (2004). Activated glia induce neuron death via MAP
Singhto, N., Sintiprungrat, K., & Thongboonkerd, V. (2013). Alterations in macrophage cel- kinase signaling pathways involving JNK and p38. Glia 45, 170–179.
lular proteome induced by calcium oxalate crystals: The association of HSP90 and F- Xiong, X. Y., Liu, L., & Yang, Q. W. (2016). Functions and mechanisms of microglia/macro-
actin is important for phagosome formation. Journal of Proteome Research 12, phages in neuroinflammation and neurogenesis after stroke. Progress in Neurobiology
3561–3572. 142, 23–44.
Son, S. M., Jung, E. S., Shin, H. J., Byun, J., & Mook-Jung, I. (2012). Aβ-induced formation of Xu, K., & Geczy, C. L. (2000). IFN-gamma and TNF regulate macrophage expression of the
autophagosomes is mediated by RAGE-CaMKKβ-AMPK signaling. Neurobiology of chemotactic S100 protein S100A8. Journal of Immunology 164, 4916–4923.
Aging 33, 1006.e11–1006.e23. Xu, K., Yen, T., & Geczy, C. L. (2001). IL-10 up-regulates macrophage expression of the
Song, B. J., Akbar, M., Abdelmegeed, M. A., Byun, K., Lee, B., Yoon, S. K., et al. (2014). Mito- S100 protein S100A8. Journal of Immunology 166, 6358–6366.
chondrial dysfunction and tissue injury by alcohol, high fat, nonalcoholic substances Yamagishi, S., & Matsui, T. (2010). Advanced glycation end products, oxidative stress and
and pathological conditions through post-translational protein modifications. Redox diabetic nephropathy. Oxidative Medicine and Cellular Longevity 3, 101–108.
Biology 3, 109–123. Yamagishi, S., Nakamura, K., Matsui, T., & Takeuchi, M. (2006). Minodronate, a nitrogen-
Song, J. S., Kang, C. M., Park, C. K., Yoon, H. K., Lee, S. Y., Ahn, J. H., et al. (2011). Inhibitory containing bisphosphonate, is a promising remedy for treating patients with diabetic
effect of receptor for advanced glycation end products (RAGE) on the TGF-β-induced retinopathy. Medical Hypotheses 66, 273–275.
alveolar epithelial to mesenchymal transition. Experimental & Molecular Medicine 43, Yammani, R. R., Long, D., & Loeser, R. F. (2009). Loeser interleukin-7 stimulates secretion
517–524. of S100A4 by activating the JAK-STAT signaling pathway in human articular
Srikanth, V., Maczurda, A., Phan, T., Steele, M., Westcott, B., Juskiw, D., et al. (2011). Ad- chondrocytes. Arthritis and Rheumatism 60, 792–800.
vanced glycation endproducts and their receptor RAGE in Alzheimer's disease. Yan, S. F., D'Agati, V., Schmidt, A. M., & Ramasamy, R. (2007). Receptor for advanced
Neurobiology of Aging 32, 763–777. glycation end products (RAGE): A formidable force in the pathogenesis of the cardio-
Steinhoff, M., Buddenkotte, J., Shpacovitch, V., Rattenholl, A., Moormann, C., Vergnolle, N., vascular complications of diabetes & aging. Current Molecular Medicine 7, 699–710.
et al. (2005). Proteinase-activated receptors: Transducers of proteinase-mediated sig- Ye, C., Choi, J. G., Abraham, S., Wu, H., Diaz, D., Terreros, D., et al. (2012). Human macro-
naling in inflammation and immune response. Endocrine Reviews 26, 1–43. phage and dendritic cell-specific silencing of high-mobility group protein B1 amelio-
Stern, D., Yan, S. D., Yan, S. F., & Schmidt, A. M. (2002). Receptor for advanced glycation rates sepsis in a humanized mouse model. Proceedings of the National Academy of
endproducts: A multiligand receptor magnifying cell stress in diverse pathologic set- Sciences of the United States of America 109, 21052–21057.
tings. Advanced Drug Delivery Reviews 54, 1615–1625. Yoo, Y., Byun, K., Kang, T., Bayarsaikhan, D., Kim, J. Y., Oh, S., et al. (2015). Amyloid-beta-
Sugihara, Y., Nojima, H., Matsuda, H., Murakami, T., Yoshikawa, M., & Kimura, I. (2000). activated human microglial cells through ER-resident proteins. Journal of Proteome
Antihyperglycemic effects of gymnemic acid IV, a compound derived from Gymnema Research 14, 214–223.
sylvestre leaves in streptozotocin-diabetic mice. Journal of Asian Natural Products Yoshida, T., Yamagishi, S., Nakamura, K., Matsui, T., Imaizumi, T., Takeuchi, M., et al.
Research 2, 321–327. (2006). Telmisartan inhibits AGE-induced C-reactive protein production through
Takeuchi, M., & Yamagishi, S. (2004). TAGE (toxic AGEs) hypothesis in various chronic downregulation of the receptor for AGE via peroxisome proliferator-activated recep-
diseases. Medical Hypotheses 63, 449–452. tor-gamma activation. Diabetologia 49, 3094–3099.
Takeuchi, M., Takino, J. I., & Yamagishi, S. I. (2010). Involvement of the toxic AGEs (TAGE)- Zafar, H. (2012). Inhibition of protein glycation and advanced glycation end products by
RAGE system in the pathogenesis of diabetic vascular complications: A novel thera- ascorbic acid. African Journal of Biotechnology 11, 51.
peutic strategy. Current Drug Targets 11, 1468–1482. Zeng, S., Dun, H., Ippagunta, N., Rosario, R., Zhang, Q. Y., Lefkowitch, J., et al. (2009). Recep-
Tam, H. L., Shiu, S. W., Wong, Y., Chow, W. S., Betteridge, D. J., & Tan, K. C. (2010). Effects of tor for advanced glycation end product (RAGE)-dependent modulation of early
atorvastatin on serum soluble receptors for advanced glycation end-products in type growth response-1 in hepatic ischemia/reperfusion injury. Journal of Hepatology 50,
2 diabetes. Atherosclerosis 209, 173–177. 929–936.
Tang, Y. (2014). Curcumin eliminates the effect of advanced glycation endproducts Zetterström, C. K., Jiang, W., Wähämaa, H., Ostberg, T., Aveberger, A. C., Schierbeck, H.,
(AGEs) on the divergent regulation of gene expression of receptors of AGEs by et al. (2008). Pivotal advance: Inhibition of HMGB1 nuclear translocation as a mech-
interrupting leptin signaling. Laboratory Investigation 94, 503–516. anism for the anti-rheumatic effects of gold sodium thiomalate. Journal of Leukocyte
Teismann, P., Sathe, K., Bierhaus, A., Leng, L., Martin, H. L., Bucala, R., et al. (2012). Receptor Biology 83, 31–38.
for advanced glycation endproducts (RAGE) deficiency protects against MPTP toxici- Zhang, Z., Zhang, L., Zhou, C., & Wu, H. (2014). Ketamine inhibits LPS-induced HGMB1 re-
ty. Neurobiology of Aging 33, 2478–2490. lease in vitro and in vivo. International Immunopharmacology 23, 14–26.
Terada, C., Yoshida, A., Nasu, Y., Mori, S., Tomono, Y., Tanaka, M., et al. (2011). Gene ex- Zhou, H., Ji, X., Wu, Y., Xuan, J., Qi, Z., Shen, L., et al. (2014). A dual-role of Gu-4 in sup-
pression and localization of high-mobility group box chromosomal protein-1 pressing HMGB1 secretion and blocking HMGB1 pro-inflammatory activity during in-
(HMGB-1) in human osteoarthritic cartilage. Acta Medica Okayama 65, 369–377. flammation. PloS One 9, e89634.
K. Byun et al. / Pharmacology & Therapeutics 177 (2017) 44–55 55

Zhou, Y., Wang, Y., Kovacs, M., Jin, J., & Zhang, J. (2005). Microglial activation induced by Zhu, T., Wang, D. X., Zhang, W., Liao, X. Q., Guan, X., Bo, H., et al. (2013). Andrographolide
neurodegeneration: A proteomic analysis. Molecular & Cellular Proteomics 4, protects against LPS-induced acute lung injury by inactivation of NF-κB. PloS One 8,
1471–1479. e56407.
Zhu, Y., Chen, X., Pan, Q., Wang, Y., Su, S., Jiang, C., et al. (2015). A comprehensive prote- Ziskoven, C., Jäger, M., Zilkens, C., Bloch, W., Brixius, K., & Krauspe, R. (2010). Oxidative
omics analysis reveals a secretory path-and status-dependent signature of exosomes stress in secondary osteoarthritis: From cartilage destruction to clinical presentation?
released from tumor-associated macrophages. Journal of Proteome Research 14, Orthopedic Reviews (Pavia) 2, e23.
4319–4331.
Zhu, X., Raina, A. K., Lee, H. G., Casadesus, G., Smith, M. A., & Perry, G. (2004). Oxidative
stress signalling in Alzheimer's disease. Brain Research 1000, 32–39.

You might also like