ISSN: 2455-8583
Global Journal of Obesity, Diabetes and Metabolic Syndrome
Review Article       Open Access      Peer-Reviewed

Role of Advanced Glycation End Products in the Progression of Diabetes Mellitus

Hengli Guo1 and Youhua Xu1,2*

1Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao
2State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Avenida Wai Long, Taipa, Macao
*Corresponding author: Dr. Youhua Xu, Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, Tel: +853-88972452; Fax: +853-28825123; Email: yhxu@must.edu.mo
Received: 08 February, 2017 | Accepted: 21 February, 2017 | Published: 22 February, 2017
Keywords: Advanced glycation end products; Diabetes mellitus; Hyperglycemia memory; Rage

Cite this as

Guo H, Xu Y (2017) Role of Advanced Glycation End Products in the Progression of Diabetes Mellitus. Glob J Obes Diabetes Metab Syndr 4(1): 024-035. DOI: 10.17352/2455-8583.000019

Diabetes Mellitus (DM) has become a world problem that seriously affected quality of life in concerned population; however, studies concerning its etiology and therapeutics are not so satisfactory. Hyperglycemia and oxidative stress damage are two hallmarks that aggravate the progression of each other. During this process, there will generate amounts of by-products, among which advanced glycation end products (AGEs) have been demonstrated to play a pivotal role in promoting the beginning and progression of DM. AGEs may interact with its receptor named RAGE and induce a series of pathological effects, such as oxidative stress, apoptosis, and inflammation etc., and form the so-called “hyperglycemia memory”. This article aims to review the pivotal role of AGEs in the progression of DM..

Introduction

Due to the population aging, urbanization and associated lifestyle changes, the incidence of diabetes worldwide is dramatically increased [1]. It is reported that the number of diabetic population has doubled over the past three decades [2] and this number will increase to 591.9 million by 2035 [3].

Diabetes is mainly composed of two types, namely type 1 and 2 diabetes mellitus (T1DM and T2DM), and T2DM shares more than 90% of diabetic population [1]. Previously, most cases of T2DM were observed in developed rather than developing countries; for instance, the prevalence of T2DM was less than 1% in China in 1980 [4]. However, this trend is completely changed at present. It is reported that 80% cases of diabetes worldwide live in developing countries or areas [5], and Asia has become a “diabetes epicenter” in the world due to its rapid economic development and western food popularization [4]. More importantly, the proportion of young cases with T2DM is higher in developing rather than in developed countries [5], strongly suggesting the association between lifestyle change and the risk of occurring with T2DM. This suggestion is supported by another fact that the global epidemic of T2DM is positively related with overweight and obesity. According to a published report, the global prevalence of overweight or obesity may reach to 57.8% in 2030 [6]. As overweight or obesity are important risk factors of T2DM [7,8], diet-associated factors may significantly exacerbate the prevalence of DM.

A range of metabolic abnormalities, in addition to hyperglycemia, are seen in the diabetes. However, it is obvious from studies in diabetic patients that glucose is the predominant metabolic abnormality and one of the major systemic risk factors in diabetes [9].

Besides hyperglycemia, Oxidative Stress (OS) is widely recognized by investigators as another hallmark and key component in the development of diabetes [10]. It is demonstrated that hyperglycemia itself can contribute to OS by forming glycation products that propagate free radicals and enlarge oxidative damage [10]. Moreover, OS itself can induce free radical damage to the DNA and finally promote cell apoptosis [11]; furthermore, OS may directly and indirectly up-regulate inflammatory proteins expression and exaggerate diabetic inflammation.

Under hyperglycemia and OS settings, a series of pro-diabetic factors will be generated, such as pro-inflammatory cytokines and advanced glycation end products (AGEs). Within this decade, more and more studies elucidated the pivotal role of AGEs in the occurrence and progression of DM. This article aims to review the role of AGEs in the development of DM.

The generation and accumulation of ages

As have been well demonstrated, level of AGEs in the body is increased during aging and may be accelerated under pathophysiological conditions such as hyperglycemia. Concerning the generation process of AGEs, there are mainly two sources responsible for the accumulation of AGEs in the body: one is external derived and the other is internal originated [12].

External-derived AGEs: Investigations have observed that AGEs can be directly derived from external sources such as heat-treated food [13]. It has been well demonstrated that foods high in protein and fat, such as meat, cheese, and egg yolk, are rich in AGEs [14]; while foods high in carbohydrates have the lowest amount of AGEs. In addition, increased cooking temperatures and times, like broiling and frying, will lead to increased amounts of AGEs [15]. In fact, diet-originated AGEs can be directly absorbed through the gut [16]. It is observed that diet heavy in AGEs will result in proportional elevations in serum AGEs level and increased AGEs cross-linking in diabetic patients [17]. On the contrary, strictly restriction of dietary AGEs can dramatically decrease serum AGEs level by as high as 30% ~ 40% [15].

After absorption, dietary AGEs can also mediate AGEs-related tissue injury like internal-originated AGEs. For instance, a study found that diabetic patients on a high-AGEs diet have increased expression and activity of MAPK, NF-κB, and VCAM-1 compared with diabetic subjects on a low-AGE diet [18]; while restricting dietary AGEs intake demonstrates decreased circulating AGEs and CRP levels [19] and suppressed AGEs-related tissue injury [20].

Concerning controlling the content of external-originated AGEs, investigations show a possibility to decrease the production of AGEs. By administration with aminoguanidine (an inhibitor of AGE formation) or AGE-albumin, scholars found in streptozotocin-induced diabetic rats that AGEs-promoted vascular dysfunction was ameliorated [21]. Another study from Kihara and colleagues [22] also observed that the blockade of AGE formation by aminoguanidine was help to improve neural signal transmission in diabetic rats. However, to our knowledge, there is still no satisfactory agent applied in clinic till now. It is expected that exploring such agents may open a novel market toward DM intervention.

Internal-originated AGEs: As have been well demonstrated, advanced glycation and oxidation increases during aging and these processes are accelerated under pathophysiological conditions such as hyperglycemia. McPherson and colleagues [23] found the intracellular AGE formation is significantly increased in endothelial cells after 1 week in a hyperglycemic environment. Thus, AGEs accumulated during aging may function as persistent “endogenous danger signals” that set the stage for the manifestation of various complications in older persons [24, 25].

Reducing sugars including glucose, fructose, and trioses can react non-enzymatically with the amino groups of proteins to form reversible Schiff bases and then Amadori products. These early glycation products undergo further complex reactions such as rearrangement, dehydration and condensation to become irreversibly cross-linked heterogeneous fluorescent derivatives termed advanced glycation end products (AGEs) [26], and scholars defined this AGEs formation process as “Maillard reaction”.

Specifically, Maillard reaction begins from Schiff bases and the Amadori product, a 1-amino-1-deoxyketose, produced by the reaction of the carbonyl group of a reducing sugar with proteins, lipids, and nucleic acid aminos [27,28]. During Amadori reorganization, these highly reactive intermediate carbonyl groups, known as alpha-dicarbonyls or oxoaldehydes, accumulate and induce corresponding damage to the tissue [10,28,29] and this process is called “carbonyl stress.” It is reported that the alpha-dicarbonyls will react with amino, sulfhydryl, guanidine, lysine and arginine functional groups in proteins and results in the denaturation and cross-linking of the involved proteins [30-32], and finally leads to the formation of stable AGE compounds [32]. Several lines of evidence indicate that accumulation of these reactive carbonyl intermediates is consequent upon hyperglycemia in diabetes. Factors responsible for the formation of AGEs include the rate of turnover of proteins for glycoxidation, the degree of hyperglycemia and oxidant stress in the environment; and if one or more of these conditions is present, both intracellular and extracellular proteins may be glycated and oxidized [28,33,34].

So far, only a few AGEs structures have been identified in vivo, such as Nε-(carboxymethyl) lysine (CML), pentosidine, imidazolones, and oxalic acid monolysinylamide (OMA) et al. [35]. Studies found these Maillard products will accumulate throughout the body, and vascular tissue is the most deeply affected one [36-38]. Although there is evidence that enzymes, such as glyoxalase-1, may detoxify AGE precursors thus inhibit AGE production [39], it should be noted that once AGEs are formed, they are nearly irreversible [40].

The progression of DM has been demonstrated to be closely accompanied with the generation process of AGEs. In this sense, comprehending the pathophysiological role of AGEs in DM is help to understand DM in both clinical practice and pharmacological studies. Previous studies indicate that AGE proteins prepared in vitro possess similar cross-reactive AGE epitopes that are common to proteins modified by AGE in vivo [41]. Therefore, both the endogenous formed AGEs and the exogenous derived AGEs, over-cooking foods for example, would damage the human bodies.

Role of age in DM

In the year 1980s, Brownlee and colleagues [26,42,43] firstly described the harmful consequences of AGE formation on the cardiovascular and renal systems in human and diabetic rats. Studies have confirmed that AGE cross-links accumulate in diabetic patients and animals and will cause a series of diabetic complications [44].

As discussed above, AGEs cross-links are permanent and irreversible complexes formed when glucoses bind to the target proteins. A bad news is that these target proteins are usually housekeeping proteins including collagen and elastin, which play an integral role in the maintenance of tissue integrity [45]. Therefore, once AGEs are formed, they will remain and continuously damage the tissue until the proteins involved are degraded [46]. General mechanisms through which AGEs contribute to diabetic complications include (1) formation of cross-links in AGEs and (2) interaction of AGEs with RAGE on cell surfaces.

Oxidative Stress-related Hyperglycemic Memory

Oxidative Stress and DM: Cells actively maintain a proper level of intracellular reactive oxygen species (ROS) through defense systems including antioxidant enzymes and low molecular weight antioxidants under physiological conditions. However, hyperglycemia-promoted over-generation of ROS will accelerate the progression of diabetic complications [9]. Several lines of evidence demonstrate that oxidative stress (OS) or ROS are involved in the pathogenesis of diabetes [10]. On the other hand, it is found that the antioxidant capacity is compromised in diabetes [47]. Altered levels of circulating antioxidants [48] and intracellular scavenging enzymes [49] provide further evidence for increased OS in diabetes. At present, OS has been widely recognized as a key component in the development of diabetes and diabetic complications [50].

Reports have shown that antioxidant therapy protects against the development of kidney disease in animal models of diabetes [47,51] and experimental evidence obtained from in vitro studies showed that prevention of ROS generation defends against the damaging effects of a hyperglycemic milieu on mesangial cell function [52]. Indeed, the overexpression of cellular antioxidants, such as copper- or zinc-containing superoxide disumtase, protect against end-organ damage in diabetic model [53].

Oxidative Stress and AGEs: AGEs, as the oxidative products, can induce OS, promote cell apoptosis, and finally induce both micro- and macro-vascular disease in diabetic patients [26, 54-57]. Direct cellular challenge with AGEs has been shown to cause OS in various cell types [58-60]. It is found that Schiff-base products and Amadori products themselves cause ROS production [61], while Nitric Oxide (NO) donors can scavenge free radicals and inhibit AGEs formation [62]. Moreover, there is a significant correlation between the levels of AGEs and antioxidant enzymes, such as glutathione peroxidase and Cu/Zn-superoxide dismutase, in uremic plasma [63]. Studies found AGE accumulation itself is a source of OS. In hyperglycemic environments, glucose can undergo auto-oxidation and generate OH radicals [61,64]. Besides that, Quagliaro and colleagues [65] observed that AGEs can stimulate production of superoxide within the cells, which may induce free radical damage to the DNA and finally cause cell apoptosis.

Oxidative Stress and Hyperglycemia Memory: Hyperglycemia is a hallmark of DM and hyperglycemia itself can promote formation of reactive oxygen species (ROS), which can interact with both DNA and proteins, and cause damage. It is found that the mitochondrial DNA may be an especially relevant target of hyperglycemia-promoted damage [66]. Xie et al. [66] found ROS mediated cellular damage is a form of pathologic “memory” in the microvasculature that persists even after glucose normalization, as suggested in human retinal vascular endothelial cells. Therefore, ROS may directly damage both islet beta-cells and insulin-sensitive cells thus lead to hyperglycemia. ROS may also link hyperglycemia with other pathways implicated in diabetic vascular complications, including AGEs formation, protein kinase (PK)C activation, increased polyol flux, and hexosamine formation [39,67].

It should be noted that hyperglycemia, AGEs and OS may facilitate the genesis of each other and constitute a vicious circle. This vicious circle promotes the formation and accumulation of AGEs in various tissues as well as in plasma at an extremely accelerated rate in diabetes [68]. On the other hand, hyperglycemia can activate nuclear factor-kappaB (NF-κB) [69], a key mediator that regulates multiple pro-inflammatory target genes. The genetic and non-genetic pathway serves each other and forms another vicious circle to promote the progression of diabetes and forms the so-called hyperglycemic memory.

Hyperglycemic memory is firstly demonstrated by study in dogs. It is reported in diabetic dogs that euglycemia could not inhibit the progression of diabetic microvascular dysfunction, thus scholars named this phenomenon as “hyperglycemic memory” [70-72]. In fact, this phenomenon also exists in human beings [73]. Two possible mechanisms are proposed that responsible for this phenomenon. One is AGEs and the other is OS. As discussed above, once AGEs are formed, they are hardly degraded. So AGEs may continuously damage blood vessels even blood glucose is lowered. On the other hand, the generation of AGEs will cause oxidative stress and oxidative stress will further promote AGEs-induced tissue damage [74-77].

Receptor-mediated Oxidative Stress: Besides direct damage, AGEs also enhance cell OS damage via a receptor-mediated pathway. Interaction of AGEs with the cell-associated RAGE is associated with ROS generation and NF-κB activation [59,60]. And AGEs-RAGE interaction leading to an increase in ROS production has been shown to cause cell apoptosis [78]. There is a study suggests that NADPH oxidase plays an important role in AGE-RAGE dependent ROS generation and gene activation [79]. The role of RAGE in the progression of DM will be discussed hereinafter this review.

Chronic low grade inflammation

The cytokine/adipokine profiles of Mexican Americans with diabetes suggest an association between low-grade inflammation and quality of glucose control [80]. It has been well demonstrated that diabetes as whole was strongly associated with elevated levels of IL-6, leptin, CRP and TNF-α, whereas worsening of glucose control was positively and linearly associated with high levels of IL-6, and leptin. Thus, low-grade inflammation is also believed as a hallmark of diabetes. Previous studies have demonstrated the role of AGEs in driving inflammatory response in macrophages [81].

AGEs-mediated Inflammatory Cell Infiltration: Clinical studies have found significant increase of AGEs accumulation in vascular tissues [82], which may induce monocyte migration across an endothelial cell monolayer [83] and promote expression of inflammatory cytokines such as interleukin (IL)-1β and TNF-α mRNA [84]. In fact, scholars have observed AGEs to be present in the plasma and to accumulate in tissues at an accelerated rate in diabetes [26,85].

AGEs also increase permeability and expression of procoagulant activity in cultured endothelium, and induce migration of mononuclear phagocytes, as well as production of platelet-derived growth factor and cytokines [86]. Schmidt et al. [87] observed that levels of AGEs and receptor for AGEs (RAGE) are increased in streptozotocin (STZ)-treated (diabetic) apolipoprotein (apo) E-null mice that have advanced atherosclerosis by 14 weeks of age. Basta and colleagues [88] further found that AGEs located on proteins, in addition to immobilized AGEs on the subendothelium, bound RAGE on the endothelium and induced hyperpermeability in diabetes. These findings are supported by another report that administration of soluble RAGE (sRAGE) inhibits vascular leakage in the intestine and skin of STZ-treated rats [34].

Adhesion Molecules in Inflammatory Cell Infiltration: Leukocyte infiltration into inflammatory lesions is mediated by sequential engagement of cell adhesion molecules and chemokines. Leukocyte influx begins with leukocyte rolling and firm attachment (sticking) to the endothelium, followed by transmigration across the endothelial surface [89]. The blockade of specific cytokines and chemokines involved in processes such as the recruitment of inflammatory cells, including monocyte MCP-1, is demonstrated to be a valid therapeutic strategy in models of diabetic nephropathy. For example, blockade of the chemokine MCP-1 could attenuate not only macrophage infiltration but also progressive renal injury in db/db mice [90].

MCP-1

Both hyperglycemia and the accumulation of AGEs can promote MCP-1 production [91, 92]. An additional influence that may induce synthesis of MCP-1 is the generation of ROS [93]. Previous reports have demonstrated that NF-κB was involved in high glucose-induced production of MCP-1 [94], it can be explained as MCP-1 promoter and enhancer regions contain NF-κB binding sequences [95]. Kislinger and colleagues investigated neuronal-associated vessels and found that RAGE is localized with its putative ligand N-epsilon-carboxymethyl lysine and NF-κB, and IL-6 [41]. It indicates the expression of MCP-1 and IL-6 is positively related with that of RAGE, and the ligation of RAGE will increase MCP-1 and IL-6 at the same time. Therefore, the expression/activation of MCP-1 and NF-κB are positively related and forms a vicious circle: the high MCP-1 contributed abundant inflammatory cells infiltration, which may release amounts of inflammatory cytokines and exacerbate inflammation status; on the other hand, inflammation status activated the intracellular NF-κB, which contributes to further production of MCP-1 and other inflammatory cytokines/proteins, such as RAGE and IL-6 etc.

ICAM-1

Besides MCP-1 as discussed above, intercellular adhesion molecule (ICAM)-1, a 90-kD cell-surface protein known as CD54 which possess five immunoglobulin-like domains, is also a major molecule involves in promoting leukocyte infiltration [92, 96]. Increased ICAM-1 expression is seen in models of type 1 [97] and type 2 diabetic nephropathy [98] in parallel with disease progression [99]. In patients with diabetes, the soluble form of ICAM-1 (sICAM-1) was observed to be elevated [100], and scholars regarded it as a powerful independent predictor of T2DM and diabetic-associated cardiovascular disease [101].

It is recognized that ICAM-1 may be even more important in promoting nephropathy associated with T2DM because its expression is not only induced by factors common to both types of diabetes, such as hyperglycemia [102], AGEs [103], hyperfiltration [104], and OS [105], but it can also be increased by additional elements characteristic of T2DM, including hyperlipidemia [106], hyperinsulinemia [107], and elevated levels of circulating TNF-α [108], which are associated with obesity [109]. In fact, the up-regulation of ICAM-1 can be observed soon after the induction of diabetes in streptozotocin (STZ)-induced diabetic rats [92, 110].

Although it is still controversial whether hyperglycemia per se can induce the expression of ICAM-1 on vascular endothelial cells, recent studies have suggested several possible mechanisms of ICAM-1 induction in diabetes: (1) ICAM-1 is induced by inflammatory cytokines such as TNF-α, IL-1, and interferon-gamma [111]; (2) Activation of protein kinase C results in upregulation of ICAM-1 on endothelial cells [112]; (3) AGEs enhance the expression of cell adhesion molecules, including ICAM-1 on vascular endothelial cells [113,114]; (4) Shear stress could also stimulate the induction of ICAM-1 [115]. Reports indicated [97,116] that shear stress, which is increased by hyperfiltration of glomerular endothelial cells, is one of the mediators of ICAM-1 induction in glomeruli; (5) OS has been documented to increase ICAM-1 expression [117]; (6) Osmotic agents up-regulate ICAM-1 expression in HUVECs [118].

Apoptosis

Most recently, there is a research demonstrated that AGEs can directly induce glomerular mesangial cells apoptosis in a dose-dependent manner [119]. In fact, the study of cell survival, or cell death (apoptosis) control, as opposed to control of mitosis, has been a focus of research over the past decade. Much of the recent understanding concerns the molecular regulation of cell survival or death by the Bcl-2 (B cell lymphoma) multigene family [11,120]. The balance of Bcl-2 family proteins can protect cell from apoptosis by reducing cell OS [11,120]; according to the report from Hockenbery and colleagues [120] , Bcl-2 polypeptides localize to intracellular sites including membranes of the mitochondria, nuclei and endoplasmic reticulum where ROS are generated, and these polypeptides can inhibit apoptosis via an anti-oxidant pathway.

In fact, studies on the intracellular cross-talk between ROS and apoptosis are abundant and have been well demonstrated. One of these pathways involves MAP kinases (p38MAPK, extracellular signal-regulated kinase1/2 (ERK1/2), and JNK etc.). Reports demonstrated that the activation of p38MAPK involves AGEs-induced fibroblast apoptosis [78] and podocyte apoptosis [121]. And another pathway involves PI3K-Akt. Akt is a cell survival factor. Growth factors or extracellular signals lead to the activation of phosphoinositide kinase, which results in Akt phosphorylation and activation leading to cell proliferation. Therefore, Akt activation by phosphorylation is an antiapoptotic signal that may protect cells from programmed cell death and promote survival [122]. Further studies found that Forkhead transcription factors (FOXOs) may participate in Akt-mediated cell survival. FOXOs, including FOXO1, FOXO3, and FOXO4, are a family of proteins that function as sensors of signaling pathways and modulate apoptosis, cell cycle, and metabolism through regulation of gene expression [123]. Akt regulates FOXO activity by inducing a prompt and sustained nuclear exclusion. There is broad consensus on the fact that Akt-dependent phosphorylation is crucial to the regulation of FOXO function. Kops et al. [124] observed activated-Akt phosphorylates and inactivates FOXO proteins. In the absence of Akt inhibition, FOXO is translocated to the nucleus leading to gene activation. FOXO transcription factors activate three major groups of genes: anti-oxidant genes, cell cycle arrest genes, and apoptotic genes [125]. It is reported that AGEs activate FOXO4 leading to apoptosis in podocytes [121] and FOXO1 leading to apoptosis in fibroblasts [78]. Further studies found the activation of JNK also plays a role in in FOXO activity [126,127].

Receptors for age

Mechanisms involved in the pathophysiological role of AGEs in diseases mainly lies in two aspects: (1) oxidative stress damage, which is directly induced by AGEs and has been discussed above; and (2) receptor mediated injury.

Types of AGEs receptors

Amounts of studies indicate that most of AGE-modified tissue damage is via AGE receptors. AGE receptors are found in a number of different cell types [128]. To now, a series of AGEs receptors have been identified and the most important receptor type is named as “the receptor for AGE (RAGE)” [40]. The other receptor types include (1) oligosaccharyltranferase-48, also called AGE-R1 [128]; (2) 80K-H, known as AGE-R2 [128]; (3) galectin 3, known as AGE-R3 [129]; (4) lactoferrin-like AGE binding protein [130]; (4) scavenger receptors (SR) class A, type II (MSR-AII) [131] and (5) SR class B, type I (MSR-BI, also known as CD36) [132].

The AGE-receptor system can be divided into two arms: one is associated with increased OS, growth, and inflammatory effects, best represented by RAGE [103]; and the other, involved in the clearance and possible detoxification of AGEs [133], includes transduce cellular signals after engagement by AGEs [133]. In this sense, although several types of AGEs receptors exist, only RAGE is demonstrated to mediate its harmful effects [40].

Structure of RAGE

The receptor for AGEs (RAGE) is the first one that described as a receptor for the products of nonenzymatic glycation and oxidation of proteins. Thereafter, besides AGEs, amounts of natural ligands of RAGE are found, including high mobility group box 1 (HMGB1), S100/calgranulins, Mac-1, phosphatidylserine and amyloid (Aβ) can also serve as RAGE ligands [134,135]. Like AGEs, an increased level of RAGE had been found in cardiac and renal tissue in diabetes [136].

RAGE is a member of the immunoglobulin superfamily [137]. The human RAGE gene is on Chromosome 6 in the major histocompatibility complex between genes for class II and class III. RAGE has a single transmembrane domain followed by a highly charged 43–amino acid cytosolic tail [138]. Specifically, it has a 332–amino acid extracellular component, consisting of two “C”-type domains preceded by one “V”-type immunoglobulin-like domain. The V domain in the N-terminus has two Nglycosylation sites and is responsible for most (but not all) extracellular ligand binding [139], and the cytoplasmic tail is believed to be essential for intracellular signaling, possibly binding to diaphanous-1 to mediate cellular migration [140]. RAGE is expressed as both full-length, membrane-bound form (fl-RAGE or mRAGE) and various soluble forms (sRAGE) which lack the transmembrane domain. While a form of RAGE that lacks the cytosolic tail but stays embedded in the membrane where it binds AGEs functions as a dominant-negative RAGE, unable to transduce a cell signal on ligand engagement.

During homeostasis, most of the tissues express a basal level of RAGE [136]. As a member of the immunoglobulin (Ig) superfamily protein, RAGE is low expressed in normal tissue and vasculature [64] and this basal level of RAGE expression involves in embryonic growth and development [141], cellular proliferation and survival [142] and the activation of various signaling events [143].

Tissues do not express significant amounts of RAGE under physiological conditions but can be induced to express RAGE in situations where either ligand accumulate and/or various transcription factors regulating RAGE are activated [144]. AGEs activation of RAGE is found in diabetes, neurodegeneration, and aging [145]. Collison and colleagues [146] demonstrated that RAGE will bind to AGE-modified albumin but not nonglycated albumin. In the diabetic vasculature, cells expressing high levels of RAGE are often proximal to or colocalized in areas where AGEs are abundant [147].

Oxidative stress promoted RAGE expression

RAGE expression is found to be elevated in high-OS states including diabetes [147], which implies a sustained ROS and AGEs generation, resulting in RAGE activation, and so on. This cycle could conceivably alter the cell’s phenotype, obscuring other receptor properties or depleting cellular antioxidant systems [148]. Furthermore, the activation of RAGE has been demonstrated to engage critical signaling pathways linked to pro-inflammatory responses, resulting in activation of various inflammatory genes [149].

Role of RAGE in diabetic-inflammation

Experimental evidence and observation strongly suggest that RAGE signaling results in profound inflammation [150]. Engagement of RAGE by the ligands activates key signal transduction pathways, such as p21ras, ERK1/2 kinases and NF-κB in endothelial cells (ECs), monocytes, and vascular smooth muscle cells. Activation of RAGE by its ligands results in a “feed forward” signaling mode that upregulates RAGE expression and amplifies the cell signals. This cascade of events leads to RAGE-mediated-enhanced expression of proinflammatory mediators as demonstrated by suppression of the effects of these modified adducts in the presence of blocking antibodies to RAGE, sRAGE, the extracellular ligand-binding domain of the receptor, or by transient transfection of cDNA encoding cytosolic tail-deleted RAGE into RAGE-bearing cells [41,130,151]. One mechanism underlying this observation probably is the presence of at least two functional binding elements for NF-κB in the promoter of the gene encoding RAGE [152,153]. In diabetic apolipoprotein E (apo E) deficient mice, RAGE signaling mediates prolonged vascular inflammation, and enhances the expression of vascular cell adhesion molecule (VCAM)-1 and tissue factor, leading to an exacerbation of the inflammatory state [154]. In addition, RAGE participates in diabetes associated atherosclerosis [155]. The role of RAGE as an inflammatory mediator is further demonstrated by application of RAGE antibodies or genetic knock-out methods. A report found that soluble RAGE (sRAGE) may play as a direct inhibitor of leukocyte recruitment [156].

By functioning as a co- or counter-receptor of the adhesion molecules, RAGE facilitates the recruitment of leukocytes to the injured vascular tissues. Amounts of reported studies have demonstrated that ligand-activated RAGE serves as an adhesion receptor that interacts with integrins and facilitates the recruitment of proinflammatory leukocytes to the sites of inflammation, and further enhancing the inflammatory state [157]. Some scholars also recognized RAGE plays as a counter-receptor for leukocyte integrins and directly contributs to the recruitment of inflammatory cells in vivo and in vitro. Pullerits et al. [158] reported that RAGE acts as an endothelial adhesion receptor that mediates interactions with the β2 integrin Mac-1. Evidence suggests neutrophils and myelomonocytic cells adhere to immobilized RAGE or RAGE-transfected cells, and this interaction is attributed to Mac-1 interactions [159]. Such interactions are augmented by the addition of S100B ligand. Hence, this RAGE-Mac-1-dependent leukocyte recruitment may be involved in the ICAM-1-independent leukocyte transmigration as observed in the ICAM-1 deficient mice [160]. But some scholars believe that RAGE may serve as an “indirect” promoter in inflammatory cell recruitment because RAGE mediated cellular activation and upregulation of adhesion molecules and proinflammatory factors [156]. Whatever, it seems that RAGE plays a significant role in the process of inflammatory cell adhesion and infiltration. Studies demonstrated that RAGE is highly expressed in macrophages, T lymphocytes, and B lymphocytes [161], and this expression may contribute to the inflammatory mechanisms.

Another impact of RAGE over-expression on diabetic inflammation is that the activation of RAGE by AGEs increases endothelial permeability to macromolecules [152]. Basta and colleagues [88] found that AGEs located on proteins, in addition to immobilized AGEs on the subendothelium, bind RAGE on the endothelium and induce hyperpermeability in diabetes. This finding is supported by another report than administration of sRAGE inhibits vascular leakage in the intestine and skin of streptozotocin-treated rats [34]. Reports have demonstrated that RAGE expression can be up-regulated on AGEs accumulation [64] and limiting RAGE expression can down-regulate expression of pro-inflammatory cytokines and attenuate vasculitis development [162]. Converging with the above discussions, downregulating RAGE should have significant role in protecting the organism against AGEs-induced inflammatory damage.

Due to the significant role of RAGE during the development of diabetes, blockade of RAGE ligation has been regarded as an effective way towards AGEs-induced diseases [43]. Because all soluble RAGE (sRAGE) species contain an intact N-terminal portion that encompasses the entire Ig-like domains, they maintain the full capacity of ligand binding, yet are devoid of signaling functions. These properties confer upon sRAGEs the status of a natural decoy for the plasma membrane-anchored RAGE [163]. As sRAGE blocks AGEs from binding to RAGE, as if sRAGE was a “sponge” soaking up soluble AGEs (sAGEs) [164], it becomes a potential therapeutic tool for the treatment of inflammatory diseases including diabetes and cardiovascular diseases. Reports indicated that RAGE participates in the formation of diabetes associated atherosclerosis in RAGE/apolipoprotein E double knock mice [155]. Another study observed that blockade of RAGE with anti-RAGE IgG or sRAGE inhibited NF-κB activation [34]. Furthermore, study found that the vascular inflammatory phenotypes such as accelerated expression of VCAM-1, tissue factor, or matrix metalloproteinases in mice are also prevented by sRAGE treatment [165]. Since AGEs may also form adducts with extracellular matrix (ECM), they may affect the structural integrity of the vessel in addition to signaling via RAGE [166]. Thus, sRAGE may also prevent such “non-specific” adverse effects of AGEs, especially during aging. Despite of its benefits on preventing RAGE ligation, sRAGE cannot be widely applied clinically due to its PROTEIN nature, which may induce immunogenicity in vivo.

Conclusion

The global prevalence of diabetes has seriously influenced the quality of life as a whole, and diabetes and diabetic dramatically increase the financial burden of the health system worldwide. Hyperglycemia and oxidative stress are two hallmarks of diabetes, and the generation of AGEs has been demonstrated to be accompanied with hyperglycemia and oxidative stress. AGEs can promote the development of diabetes via both direct and indirect pathway. On the one hand, AGEs augmented oxidative stress; on the other hand, AGEs active RAGE and induce receptor-mediated cell damage, such as apoptosis and inflammation.

The intervention of AGEs-involved damage should help to ameliorate the development of diabetes. Currently, there are mainly two strategies can be considered. Firstly, AGEs oriented strategy. Either reducing the intake of AGEs-rich foods or using AGEs formation inhibitor aminoguanidine can directly reduce AGEs-oriented damage to the organism. The second strategy if receptor oriented. Application of soluble RAGE (sRAGE) or inhibiting expression of RAGE has been demonstrated to possess effects on ameliorating diabetic complications. However, there is still no satisfactory agents or drugs applied in clinic that target AGEs. Investigations in this area may bring a novel strategy towards treatment of diabetes and diabetic complications.

This work was supported by Science and Technology Development Fund of Macau (FDCT: 071/2014/A).

  1. Chen L, Magliano DJ, Zimmet PZ (2011) The worldwide epidemiology of type 2 diabetes mellitus-present and future perspectives. Nat Rev Endocrinol 8: 228-236. Link: https://goo.gl/4l6TfE
  2. Danaei G, Finucane MM, Lu Y, Singh GM, Cowan MJ, et al. (2011) National, regional, and global trends in fasting plasma glucose and diabetes prevalence since 1980: systematic analysis of health examination surveys and epidemiological studies with 370 country-years and 2.7 million participants. Lancet 378: 31-40. Link: https://goo.gl/OuiqjU
  3. Guariguata L, Whiting DR, Hambleton I, Beagley J, Linnenkamp U, et al. (2014) Global estimates of diabetes prevalence for 2013 and projections for 2035. Diabetes Res Clin Pract 103: 137–149. Link: https://goo.gl/s6m52B
  4. Chan JC, Malik V, Jia W, Kadowaki T, Yajnik CS, et al. (2009) Diabetes in Asia: epidemiology, risk factors, and pathophysiology. JAMA 301: 2129-2140. Link: https://goo.gl/ufuhX2
  5. Shaw JE, Sicree RA, Zimmet PZ (2010) Global estimates of the prevalence of diabetes for 2010 and 2030. Diabetes Res Clin Pract 87: 4-14. Link: https://goo.gl/4STa6z
  6. Kelly T, Yang W, Chen CS, Reynolds K, He J (2008) Global burden of obesity in 2005 and projections to 2030. Int J Obes (Lond) 32: 1431-1437. Link: https://goo.gl/QMyyg1
  7. Hu FB, Manson JE, Stampfer MJ, Colditz G, Liu S, et al. (2001) Diet, lifestyle, and the risk of type 2 diabetes mellitus in women. N Engl J Med 345: 790-797. Link: https://goo.gl/j6TnXI
  8. Narayan KM, Boyle JP, Thompson TJ, Gregg EW, Williamson DF (2007) Effect of BMI on lifetime risk for diabetes in the U. S. Diabetes Care 30: 1562-1566. Link: https://goo.gl/ri88eJ
  9. Sourris KC, Forbes JM, Cooper ME (2008) Therapeutic Interruption of Advanced Glycation in Diabetic Nephropathy. Ann N Y Acad Sci 1126: 101-106. Link: https://goo.gl/F8GbL4
  10. Baynes JW, Thorpe SR (1999) Role of oxidative stress in diabetic complications: a new perspective on an old paradigm. Diabetes 48: 1-9. Link: https://goo.gl/AiCibg
  11. Xu Y, Feng L, Wang S, Zhu Q, Lin J (2011) Phytoestrogen calycosin-7-O-β-D-glucopyranoside ameliorates advanced glycation end products-induced HUVEC damage. J Cell Biochem 112: 2953-2965. Link: https://goo.gl/9dDdRU
  12. Garg D, Merhi Z (2005) Advanced Glycation End Products: Link between Diet and Ovulatory Dysfunction in PCOS? Nutrients 7: 10129–10144. Link: https://goo.gl/oTxHBN
  13. Uribarri J, Cai W, Peppa M, Goodman S, Ferrucci L (2007)  Circulating glycotoxins and dietary advanced glycation endproducts: two links to inflammatory response, oxidative stress, and aging. J Gerontol A Biol Sci Med Sci 62: 427-433. Link: https://goo.gl/H5WvFt
  14. Vlassara H, Cai W, Crandall J, Goldberg T, Oberstein R (2002) Inflammatory mediators are induced by dietary glycotoxins, a major risk factor for diabetic angiopathy. Proc Natl Acad Sci U S A 99: 15596 -155601. Link: https://goo.gl/KnH30F
  15. Goldberg T, Cai W, Peppa M, Dardaine V, Baliga BS, et al. (2004) Advanced glycoxidation end products in commonly consumed foods. J Am Diet Assoc 104: 1287-1291. Link: https://goo.gl/CVtdDN
  16. Leung C, Herath CB, Jia Z, Andrikopoulos S, Brown BE, et al. (2016) Dietary advanced glycation end-products aggravate non-alcoholic fatty liver disease. World J Gastroenterol 22: 8026-8040. Link: https://goo.gl/DtblMH
  17. Koschinsky T, He CJ, Mitsuhashi T, Bucala R, Liu C, et al. (1997) Orally absorbed reactive glycation products (glycotoxins): an environmental risk factor in diabetic nephropathy. Proc Natl Acad Sci U S A 94: 6474-6479. Link: https://goo.gl/CVjZ09
  18. Cai W, He JC, Zhu L, Peppa M, Lu C, et al. (2004) High levels of dietary advanced glycation end products transform low-density lipoprotein into a potent redox-sensitive mitogen-activated protein kinase stimulant in diabetic patients. Circulation 110: 285-291. Link: https://goo.gl/BmpnHO
  19. Peppa M, Uribarri J, Cai W, Lu M, Vlassara H (2004) Glycoxidation and inflammation in renal failure patients. Am J Kidney Dis 43: 690-695. Link: https://goo.gl/15wpVU
  20. Uribarri J, Peppa M, Cai W, Goldberg T, Lu M, et al. (2003) Restriction of dietary glycotoxins reduces excessive advanced glycation end products in renal failure patients. J Am Soc Nephrol 14: 728-731. Link: https://goo.gl/XoaU9m
  21. H Vlassara, H Fuh, Z Makita, S Krungkrai, A Cerami, et al. (1992) Exogenous advanced glycosylation end products induce complex vascular dysfunction in normal animals: A model for diabetic and aging complications [J]. Proc Natl Acad Sci U S A 89: 12043-12047. Link: https://goo.gl/g1Xj8h
  22. Kihara M, Schmelzer JD, Poduslo JF, Curran GL, Nickander KK, et al. (1991) Aminoguanidine effects on nerve blood flow, vascular permeability, electrophysiology, and oxygen free radicals. Proc Natl Acad Sci U S A 88: 6107-6111. Link: https://goo.gl/T6jRtg
  23. McPherson JD, Shilton BH, Walton DJ (1988) Role of fructose in glycation and cross-linking of proteins. Biochemistry 27: 1901-1907. Link: https://goo.gl/RocLX3
  24. Alves M, Cunha DA, Calegari VC, Saad MJ, Boschero AC, et al. (2005) Nuclear factor- κB and advanced glycation end-products expression in lacrimal glands of aging rats. J Endocrinol 187: 159-166. Link: https://goo.gl/J218qH
  25. Simm A, Casselmann C, Schubert A, Hofmann S, Reimann A, et al. (2004) Age associated changes of AGE-receptor expression: RAGE upregulation is associated with human heart dysfunction. Exp Gerontol 39: 407-413. Link: https://goo.gl/GcgIm6
  26. Brownlee M, Cerami A, Vlassara H (1988) Advanced glycosylation end products in tissue and the biochemical basis of diabetic complications. N Engl J Med 318: 1315-1321. Link: https://goo.gl/4vRs2p
  27. Leiva GE, Naranjo GB, Malec LS (2017) A study of different indicators of Maillard reaction with whey proteins and different carbohydrates under adverse storage conditions. Food Chem 215: 410-416. Link: https://goo.gl/4l8hSC
  28. Sharma Y, Saxena S, Mishra A,  Saxena A, Natu SM (2013) Advanced glycation end products and diabetic retinopathy. J Ocul Biol Dis Infor 5: 63-69. Link: https://goo.gl/IUZdZG
  29. Suzuki D, Miyata T, Saotome N, Horie K, Inagi R, et al. (1999) Immunohistochemical evidence for an increased oxidative stress and carbonyl modification of proteins in diabetic glomerular lesions. J Am Soc Nephrol 10: 822-832. Link: https://goo.gl/R6QU3O
  30. Lo TW, Westwood ME, McLellan AC, Selwood T, Thornalley PJ (1994) Binding and modification of proteins by methylglyoxal under physiological conditions. A kinetic and mechanistic study with N alpha-acetylarginine, N alpha-acetylcysteine, and N alpha-acetyllysine, and bovine serum albumin. J Biol Chem 269: 32299 -32305. Link: https://goo.gl/vvAm0V
  31. Song R, Yang P, Wei R, Ruan G (2016) Antioxidative, Antibacterial, and Food Functional Properties of the Half-Fin Anchovy Hydrolysates-Glucose Conjugates Formed via Maillard Reaction. Molecules 21. Link: https://goo.gl/RFOvnu
  32. Niquet-Léridon C, Jacolot P, Niamba CN, Grossin N, Boulanger E, et al. (2015) The rehabilitation of raw and brown butters by the measurement of two of the major Maillard products, N(ε)-carboxymethyl-lysine and 5-hydroxymethylfurfural, with validated chromatographic methods. Food Chem 77: 361-368. Link: https://goo.gl/Nkb3Af
  33. Schmidt A M, Hasu M, Popov D, Zhang J H, Chen J, et al. (1994) Receptor for advanced glycation end products (AGEs) has a central role in vessel wall interactions and gene activation in response to circulating AGE proteins. Proc Natl Acad Sci U S A 91: 8807-8811. Link: https://goo.gl/k9OZvD
  34. Schmidt AM, Yan SD, Wautier JL, Stern D (1999) Activation of receptor for advanced glycation end products: a mechanism for chronic vascular dysfunction in diabetic vasculopathy and atherosclerosis. Circ Res 84: 489-497. Link: https://goo.gl/bvY7y2
  35. Matsumoto T, Ozawa Y, Taguchi K, Kobayashi T, Kamata K (2010) Diabetes-associated changes and role of N epsilon-(carboxymethyl)lysine in big ET-1-induced coronary vasoconstriction. Peptides 31: 346-353. Link: https://goo.gl/OjfB1U
  36. Berg TJ, Clausen JT, Torjesen PA, Dahl-Jørgensen K, Bangstad HJ, et al. (1998) The advanced glycation end product Nepsilon-(carboxymethyl)lysine is increased in serum from children and adolescents with type 1 diabetes. Diabetes Care 21: 1997-2002. Link: https://goo.gl/lwSmiY
  37. Horie K, Miyata T, Maeda K, Miyata S, Sugiyama S, et al. (1997) Immunohistochemical colocalization of glycoxidation products and lipid peroxidation products in diabetic renal glomerular lesions. Implication for glycoxidative stress in the pathogenesis of diabetic nephropathy. J Clin Invest 100: 2995-3004. Link: https://goo.gl/7dLlK8
  38. Sakata N, Imanaga Y, Meng J, Tachikawa Y, Takebayashi S, et al. (1999) Increased advanced glycation end products in atherosclerotic lesions of patients with end-stage renal disease. Atherosclerosis 142: 67-77. Link: https://goo.gl/9hUVM8
  39. Brownlee M (2001) Biochemistry and molecular cell biology of diabetic complications. Nature 414: 813-820. Link: https://goo.gl/Lompiy
  40. Schmidt AM, Yan SD, Yan SF, Stern DM (2000) The biology of the receptor for advanced glycation end products and its ligands. Biochim Biophys Acta 1498: 99-111. Link: https://goo.gl/qTFbYs
  41. Kislinger T, Fu C, Huber B, Qu W, Taguchi A, et al. (1999) Nε-(carboxymethyl)lysine adducts of proteins are ligands for receptor for advanced glycation end products that activate cell signaling pathways and modulate gene expression. J Biol Chem 274: 31740-31749. Link: https://goo.gl/8sVOOk
  42. Brownlee M, Cerami A, Vlassara H (1988) Advanced products of nonenzymatic glycosylation and the pathogenesis of diabetic vascular disease. Diabetes Metab Rev 4: 437-451. Link: https://goo.gl/4dpSu6
  43. Brownlee M, Vlassara H, Kooney A, Ulrich P, Cerami A (1986) Aminoguanidine prevents diabetes-induced arterial wall protein cross-linking. Science 232: 1629-1632. Link: https://goo.gl/V0Nnsu
  44. Ajith TA, Vinodkumar P (2016) Advanced Glycation End Products: Association with the Pathogenesis of Diseases and the Current Therapeutic Advances. Curr Clin Pharmacol 11: 118-127. Link: https://goo.gl/ZKIvDq
  45. Brüel A, Oxlund H (1996) Changes in biomechanical properties, composition of collagen and elastin, and advanced glycation endproducts of the rat aorta in relation to age. Atherosclerosis 127: 155-165. Link: https://goo.gl/761hF0
  46. Verzijl N, DeGroot J, Bank RA, Bayliss MT, Bijlsma JW, et al. (2001) Age-related accumulation of the advanced glycation endproduct pentosidine in human articular cartilage aggrecan: the use of pentosidine levels as a quantitative measure of protein turnover. Matrix Biol 20: 409-417. Link: https://goo.gl/3oJvKV
  47. Montane J, Cadavez L, Novials A (2014) Stress and the inflammatory process: a major cause of pancreatic cell death in type 2 diabetes. Diabetes Metab Syndr Obes 7: 25-34. Link: https://goo.gl/tv8aiC
  48. Yung HW, Alnæs-Katjavivi P, Jones CJ, El-Bacha T, Golic M, et al. (2016) Placental endoplasmic reticulum stress in gestational diabetes: the potential for therapeutic intervention with chemical chaperones and antioxidants. Diabetologia, 59: 2240-2250. Link: https://goo.gl/ZehZnb
  49. Reddi AS, Bollineni JS (1997) Renal cortical expression of mRNAs for antioxidant enzymes in normal and diabetic rats. Biochem Biophys Res Commun 235: 598-601. Link: https://goo.gl/OGwrMb
  50. Saeidnia S, Abdollahi M (2013) Toxicological and pharmacological concerns on oxidative stress and related diseases. Toxicol Appl Pharmacol 273: 442–455. Link: https://goo.gl/6k2Cgh
  51. Earle KA, Zitouni K, Pepe J, Karaflou M, Godbold J (2016)  Modulation of endogenous antioxidant defense and the progression of kidney disease in multi-heritage groups of patients with type 2 diabetes: PRospective EValuation of Early Nephropathy and its Treatment (PREVENT). J Transl Med 14: 234. Link: https://goo.gl/ydla0z
  52. Studer RK, Craven PA, DeRubertis FR (1997) Antioxidant inhibition of protein kinase C-signaled increases in transforming growth factor-beta in mesangial cells. Metabolism 46: 918-925. Link: https://goo.gl/ttFBSr
  53. DeRubertis FR, Craven PA, Melhem MF (2007) Acceleration of diabetic renal injury in the superoxide dismutase knockout mouse: effects of tempol. Metabolism 56: 1256-1264. Link: https://goo.gl/A6tzMC
  54. Koike S, Yano S, Tanaka S, Sheikh AM, Nagai A, et al. (2016) Advanced Glycation End-Products Induce Apoptosis of Vascular Smooth Muscle Cells: A Mechanism for Vascular Calcification. Int J Mol Sci 17. Link: https://goo.gl/btmO3X
  55. Makita Z, Radoff S, Rayfield EJ, Yang Z, Skolnik E, et al. (1991) Advanced glycosylation end products in patients with diabetic nephropathy. N Engl J Med 325: 836-842. Link: https://goo.gl/W2Nh7e
  56. Wang Z, Li H, Guo R, Wang Q, Zhang D (2016) Antioxidants inhibit advanced glycosylation end-product-induced apoptosis by downregulation of miR-223 in human adipose tissue-derived stem cells. Sci Rep 6: 23021. Link: https://goo.gl/8l6W2f
  57. Orasanu G, Plutzky J (2009) The Pathologic Continuum of Diabetic Vascular Disease. J Am Coll Cardiol 53: S35-S42. Link: https://goo.gl/BP7Ty2
  58. Rosa CM, Gimenes R, Campos DH, Guirado GN, Gimenes C, et al. (2016) Apocynin influence on oxidative stress and cardiac remodeling of spontaneously hypertensive rats with diabetes mellitus. Cardiovasc Diabetol 15: 126. Link: https://goo.gl/klnUjJ
  59. Lander HM, Tauras JM, Ogiste JS, Hori O, Moss RA, et al. (1997) Activation of the receptor for advanced glycation endproducts triggers a MAP kinase pathway regulated by oxidant stress. J Biol Chem 272: 17810-17814. Link: https://goo.gl/fW00VB
  60. Meertens JH, Nienhuis HL, Lefrandt JD, Schalkwijk CG, Nyyssönen K, et al.  (2016) The Course of Skin and Serum Biomarkers of Advanced Glycation Endproducts and Its Association with Oxidative Stress, Inflammation, Disease Severity, and Mortality during ICU Admission in Critically Ill Patients: Results from a Prospective Pilot Study. PLoS One 11: e0160893. Link: https://goo.gl/yxyNPA
  61. Noiri E, Tsukahara H (2005) Parameters for measurement of oxidative stress in diabetes mellitus: applicability of enzymelinked immunosorbent assay for clinical evaluation. J Investig Med 53: 167-175. Link: https://goo.gl/BVZlDt
  62. Asahi K1, Ichimori K, Nakazawa H, Izuhara Y, Inagi R, et al. (2000) Nitric oxide inhibits the formation of advanced glycation end products. Kidney Int 58: 1780-1787. Link: https://goo.gl/UlTXly
  63. Ueda Y, Miyata T, Hashimoto T, Yamada H, Izuhara Y, et al. (1998) Implication of altered redox regulation by antioxidant enzymes in the increased plasma pentosidine, an advanced glycation end product, in uremia. Biochem Biophys Res Commun 245: 785-790. Link: https://goo.gl/VjYfj5
  64. Goldin A, Beckman JA, Schmidt AM, Creager MA (2005) Advanced Glycation End Products: Sparking the Development of Diabetic Vascular Injury. Circulation 114: 597-605. Link: https://goo.gl/rrWPEi
  65. Quagliaro L, Piconi L, Assaloni R, Da Ros R, Maier A, et al. (2005) Intermittent high glucose enhances ICAM-1, VCAM-1 and E-selectin expression in human umbilical vein endothelial cells in culture: the distinct role of protein kinase C and mitochondrial superoxide production. Atherosclerosis 183: 259-267. Link: https://goo.gl/C4uJVc
  66. Xie L, Zhu X, Hu Y, Li T, Gao Y, et al. (2008) Mitochondrial DNA oxidative damage triggers mitochondrial dysfunction and apoptosis in high glucose-induced human retinal vascular endothelial cells. Invest Ophthalmol Vis Sci 49: 4203-4209. Link: https://goo.gl/yFMgt2
  67. Nishikawa T, Edelstein D, Du XL, Yamagishi S, Matsumura T, et al. (2000) Normalizing mitochondrial superoxide production blocks three pathways of hyperglycaemia damage. Nature 404: 787-790. Link: https://goo.gl/OANAiw
  68. Vlassara H (1990) Chronic diabetic complications and tissue glycosylation. Diabetes Care 13: S1180-S1185. Link: https://goo.gl/gKuAKD
  69. Piga R, Naito Y, Kokura S, Handa O, Yoshikawa T (2007) Short-term high glucose exposure induces monocyte-endothelial cells adhesion and transmigration by increasing VCAM-1 and MCP-1 expression in human aortic endothelial cells. Atherosclerosis 193: 328 -334. Link: https://goo.gl/LTcPIM
  70. Engerman RL, Kern TS (1987) Progression of incipient diabetic retinopathy during good glycemic control. Diabetes 36: 808-812. Link: https://goo.gl/iJkiEk
  71. Roy S, Sala R, Cagliero E, Lorenzi M (1990) Overexpression of fibronectin induced by diabetes or high glucose: phenomenon with a memory. Proc Natl Acad Sci U S A 87: 404-408. Link: https://goo.gl/5fR4ZX
  72. Bakker W, Eringa EC, Sipkema P, van Hinsbergh VW (2009) Endothelial dysfunction and diabetes: roles of hyperglycemia, impaired insulin signaling and obesity. Cell Tissue Res 335: 165-189. Link: https://goo.gl/t6nqZC
  73. Fioretto P, Steffes MW, Sutherland DE, Goetz FC, Mauer M (1998) Reversal of lesions of diabetic nephropathy after pancreas transplantation. N Engl J Med 339: 69-75. Link: https://goo.gl/HVmH72
  74. Dworakowski R, Alom-Ruiz SP, Shah AM (2008) NADPH oxidasederived reactive oxygen species in the regulation of endothelial phenotype. Pharmacol Rep 60: 21-28. Link: https://goo.gl/FtMId4
  75. Münzel T, Sinning C, Post F, Warnholtz A, Schulz E (2008) Pathophysiology, diagnosis and prognostic implications of endothelial dysfunction. Ann Med 40: 180-196. Link: https://goo.gl/BpxQY2
  76. Rask-Madsen C, King GL (2007) Mechanisms of disease: endothelial dysfunction in insulin resistance and diabetes. Nat Clin Pract Endocrinol Metab 3: 46-56. Link: https://goo.gl/h8Qt3b
  77. Forbes JM, Coughlan MT, Cooper ME (2008) Oxidative stress as a major culprit in kidney disease in diabetes. Diabetes 57: 1446-1454. Link: https://goo.gl/1kRHWx
  78. Alikhani M, Maclellan CM, Raptis M, Vora S, Trackman PC, et al. (2007) Advanced glycation endproducts induce apoptosis in fibroblasts through activation of ROS MAP kinases and FOXO1 transcription factor. Am J Physiol Cell Physiol 292: C850-C856. Link: https://goo.gl/Qzo9IJ
  79. Wautier MP, Chappey O, Corda S, Stern DM, Schmidt AM, et al. (2001) Activation of NADPH oxidase by AGE links oxidant stress to altered gene expression via RAGE. Am J Physiol Endocrinol Metab 280: E685-E694. Link: https://goo.gl/8l0JkZ
  80. Mirza S, Hossain M, Mathews C, Martinez P, Pino P, et al. (2015) Type 2-diabetes is associated with elevated levels of TNF-alpha, IL-6 and adiponectin and low levels of leptin in a population of Mexican Americans: A cross-sectional study. Cytokine 57: 136-142. Link: https://goo.gl/aYLEKs
  81. Jin X, Yao T, Zhou Z, Zhu J, Zhang S, et al. (2015) Advanced glycation end products enhance macrophages polarization into M1 phenotype through activating RAGE/NF-B Pathway. BioMed Res Int 2015: 732450. Link: https://goo.gl/pS5f1p
  82. Soulis T, Thallas V, Youssef S, Gilbert RE, McWilliam BG, et al. (1997) Advanced glycation end products and their receptors co-localise in rat organs susceptible to diabetic microvascular injury. Diabetologia 40: 619-628. Link: https://goo.gl/MkhzpV
  83. Edelstein D, Brownlee M (1992) Mechanistic studies of advanced glycosylation end product inhibition by aminoguanidine. Diabetes 41: 26-29. Link: https://goo.gl/QLhixM
  84. Webster L, Abordo EA, Thornalley PJ, Limb GA (1997) Induction of TNF-alpha and IL-1beta mRNA in monocytes by methylglyoxal- and advanced glycated end product-modified human serum albumin. Biochem Soc Trans 25: 250S. Link: https://goo.gl/oEzV3z
  85. Dyer DG, Blackledge JA, Thorpe SR, Baynes JW (1991) Formation of pentosidine during nonenzymatic browning of proteins by glucose. Identification of glucose and other carbohydrates as possible precursors of pentosidine in vivo. J Biol Chem 266: 11654-11660. Link: https://goo.gl/LZUd6z
  86. Kirstein M, Brett J, Radoff S, Ogawa S, Stern D, et al. (1990) Advanced protein glycosylation induces transendothelial human monocyte chemotaxis and secretion of platelet-derived growth factor: role in vascular disease of diabetes and aging. Proc Natl Acad Sci U S A 87: 9010-9014. Link: https://goo.gl/Mckeoo
  87. Schmidt AM, Yan SD, Yan SF, Stern DM (2001) The multiligand receptor RAGE as a progression factor amplifying immune and inflammatory responses. J Clin Invest 108: 949-955. Link: https://goo.gl/lfOS7Q
  88. Basta G, Schmidt AM, De Caterina R (2004) Advanced glycation end products and vascular inflammation: implications for accelerated atherosclerosis in diabetes. Cardiovasc Res 63: 582-592. Link: https://goo.gl/nhbi5x
  89. Hogg N, Berlin C (1995) Structure and function of adhesion receptors in leukocyte trafficking. Immunol Today 16: 327-330. Link: https://goo.gl/8jE1WT
  90. Chow FY, Nikolic-Paterson DJ, Ma FY, Ozols E, Rollins BJ, et al. (2007) Monocyte chemoattractant protein-1-induced tissue inflammation is critical for the development of renal injury but not type 2 diabetes in obese db/db mice. Diabetologia 50: 471-480. Link: https://goo.gl/lR1Qil
  91. Ihm CG, Park JK, Hong SP, Lee TW, Cho BS, et al. (1998) A high glucose concentration stimulates the expression of monocyte chemotactic peptide 1 in human mesangial cells. Nephron 79: 33-37. Link: https://goo.gl/pLAABb
  92. Wu Y, Wu G, Qi X, Lin H, Qian H, et al. (2006) Protein kinase C beta inhibitor LY333531 attenuates intercellular adhesion molecule-1 and monocyte chemotactic protein-1 expression in the kidney in diabetic rats. J Pharmacol Sci 101: 335-343. Link: https://goo.gl/1j1BG5
  93. Schena FP, Gesualdo L, Grandaliano G, Montinaro V (1997) Progression of renal damage in human glomerulonephritides: Is there sleight of hand in winning the game. Kidney Int 52: 1439-1457. Link: https://goo.gl/Mn53UO
  94. Ha H, Yu MR, Choi YJ, Kitamura M, Lee HB (2002) Role of high glucose-induced nuclear factor-κB activation in monocyte chemoattractant protein-1 expression by mesangial cells. J Am Soc Nephrol 13: 894-902. Link: https://goo.gl/xtilql
  95. Ueda A, Okuda K, Ohno S, Shirai A, Igarashi T, et al. (1994) NF-κB and Sp1 regulates transcription of the human monocyte chemoattractant protein-1 gene. J Immunol 153: 2052-2063. Link: https://goo.gl/cxKMlw
  96. Rothlein R, Wegner C (1992) Role of intercellular adhesion molecule-1 in the inflammatory response. Kidney Int 41: 617-619. Link: https://goo.gl/zSYrX8
  97. Gong W, Chen C, Xiong F, Yang Z, Wang Y, et al. (2016) CKIP-1 ameliorates high glucose-induced expression of fibronectin and intercellular cell adhesion molecule-1 by activating the Nrf2/ARE pathway in glomerular mesangial cells. Biochem Pharmacol 116: 140-152. Link: https://goo.gl/s9YjBV
  98. Zheng X, Zhu S, Chang S, Cao Y, Dong J, et al. (2013) Protective effects of chronic resveratrol treatment on vascular inflammatory injury in streptozotocin-induced type 2 diabetic rats: Role of NF-kappa B signaling. Eur J Pharmacol S0014-2999: 00793. Link: https://goo.gl/g2Z8wr
  99. Matsui H, Suzuki M, Tsukuda R, Iida K, Miyasaka M, et al. (1996) Expression of ICAM-1 on glomeruli is associated with progression of diabetic nephropathy in a genetically obese diabetic rat, Wistar fatty. Diabetes Res Clin Pract 32: 1-9. Link: https://goo.gl/0c1iAW
  100. Becker A, van Hinsbergh VW, Jager A, Kostense PJ, Dekker JM, et al. (2002) Why is soluble intercellular adhesion molecule-1 related to cardiovascular mortality? Eur J Clin Invest 32, 1-8. Link: https://goo.gl/YmqX5W
  101. Meigs JB, Hu FB, Rifai N, Manson JE (2004) Biomarkers of endothelial dysfunction and risk of type 2 diabetes mellitus. JAMA 291: 1978-1986. Link: https://goo.gl/ZRsmCm
  102. Kanikarla-Marie P, Jain SK (2015) Role of Hyperketonemia in Inducing Oxidative Stress and Cellular Damage in Cultured Hepatocytes and Type 1 Diabetic Rat Liver. Cell Physiol Biochem 37: 2160-2170. Link: https://goo.gl/rIz7jx
  103. Chen C, Huang K, Hao J, Huang J, Yang Z, et al. (2016) Polydatin attenuates AGEs-induced upregulation of fibronectin and ICAM-1 in rat glomerular mesangial cells and db/db diabetic mice kidneys by inhibiting the activation of the SphK1-S1P signaling pathway. Mol Cell Endocrinol 427: 45-56. Link: https://goo.gl/XUyf2H
  104. Riser BL, Varani J, Cortes P, Yee J, Dame M, et al. (2001) Cyclic stretching of mesangial cells up-regulates intercellular adhesion molecule-1 and leukocyte adherence: A possible new mechanism for glomerulosclerosis. Am J Pathol 158: 11-17. Link: https://goo.gl/CK7X0d
  105. Onozato ML, Tojo A, Goto A, Fujita T (2004) Radical scavenging effect of gliclazide in diabetic rats fed with a high cholesterol diet. Kidney Int 65: 951-960. Link: https://goo.gl/ClerT4
  106. Rogil Jose de Almeida Torres, Andrea Luchini, Regiane do Rocio de Almeida Torres, Luciano Rodrigo Silva de Oliveira, Caroline Luzia de Almeida Torres, et al. (2014) Effect of candesartan on the expression of sclera-choroidal intercellular adhesion molecule-1 in hypercholesterolemic models. Clinics (Sao Paulo) 69: 145-149. Link: https://goo.gl/RM0ajk
  107. M Okouchi, N Okayama, M Shimizu, H Omi, T Fukutomi, et al. (2002) High insulin exacerbates neutrophil-endothelial cell adhesion through endothelial surface expression of intercellular adhesion molecule-1 via activation of protein kinase C and mitogen-activated protein kinase. Diabetologia 45: 556-559. Link: https://goo.gl/Q5Aydu
  108. Yokoyama H, Takaeda M, Wada T, Ohta S, Hisada Y, et al. (1997) Glomerular ICAM-1 expression related to circulating TNF-alpha in human glomerulonephritis. Nephron 76: 425-433. Link: https://goo.gl/9gNfla
  109. Hotamisligil GS, Shargill NS, Spiegelman BM (1993) Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Science 259: 87-91. Link: https://goo.gl/Ahn08C
  110. Sugimoto H1, Shikata K, Hirata K, Akiyama K, Matsuda M, et al. (1997) Increased expression of intercellular adhesion molecule-1 (ICAM-1) in diabetic rat glomeruli: glomerular hyperfiltration is a potential mechanism of ICAM-1 upregulation. Diabetes 46: 2075-2081. Link: https://goo.gl/fqxRXu
  111. Lisak RP, Bealmear B, Benjamins JA (2016) Schwann cell differentiation inhibits interferon-gamma induction of expression of major histocompatibility complex class II and intercellular adhesion molecule-1. J Neuroimmunol 295-296: 93-99. Link: https://goo.gl/GxgIla
  112. Lek HS, Morrison VL, Conneely M, Campbell PA, McGloin D, et al. (2013) The spontaneously adhesive leukocyte function-associated antigen-1 (LFA-1) integrin in effector T cells mediates rapid actin- and calmodulin-dependent adhesion strengthening to ligand under shear flow. J Biol Chem 288: 14698-14708. Link: https://goo.gl/7IjNlQ
  113. Liu Q, Qiao AM, Yi LT, Liu ZL, Sheng SM (2016) Protection of kinsenoside against AGEs-induced endothelial dysfunction in human umbilical vein endothelial cells. Life Sci 162: 102-107. Link: https://goo.gl/z3Gw5J
  114. Sengoelge G, Födinger M, Skoupy S, Ferrara I, Zangerle C, et al. (1998) Endothelial cell adhesion molecule and PMNL response to inflammatory stimuli and AGE-modified fibronectin. Kidney Int 54: 1637-1651. Link: https://goo.gl/dKQHUy
  115. Nagel T, Resnick N, Atkinson WJ, Dewey CF Jr, Gimbrone MA Jr (1994) Shear stress selectively upregulates intercellular adhesion molecule-1 expression in cultured human vascular endothelial cells. J Clin Invest 94: 885-891. Link: https://goo.gl/IvRsBL
  116. Klingberg H, Loft S, Oddershede LB, Møller P (2015) The influence of flow, shear stress and adhesion molecule targeting on gold nanoparticle uptake in human endothelial cells. Nanoscale 7: 11409-11419. Link: https://goo.gl/ZFQdkb
  117. Andrew O Odegaard, David R Jacobs Jr, Otto A Sanchez, David C Goff Jr, Alexander P Reiner, et al. (2016) Oxidative stress, inflammation, endothelial dysfunction and incidence of type 2 diabetes. Cardiovasc Diabetol 15: 51. Link: https://goo.gl/9QyGB0
  118. Taki H, Kashiwagi A, Tanaka Y, Horiike K (1996) Expression of intercellular adhesion molecules 1 (ICAM-1) via an osmotic effect in human umbilical vein endothelial cells exposed to high glucose medium. Life Sci 58: 1713-1721. Link: https://goo.gl/mHNGek
  119. Liang YJ, Jian JH, Liu YC, Juang SJ, Shyu KG, et al. (2010) Advanced glycation end products-induced apoptosis attenuated by PPARdelta activation and epigallocatechin gallate through NF-kappaB pathway in human embryonic kidney cells and human mesangial cells. Diabetes Metab Res Rev 26: 406-416. Link: https://goo.gl/A9dj4Z
  120. Hockenbery D, Nuñez G, Milliman C, Schreiber RD, Korsmeyer SJ (1990) Bcl-2 is an inner mitochondrial membrane protein that blocks programmed cell death. Nature 348: 334-336. Link: https://goo.gl/aUp7xW
  121. Chuang PY, Yu Q, Fang W, Uribarri J, He JC (2007) Advanced glycation endproducts induce podocyte apoptosis by activation of the FOXO4 transcription factor. Kidney Int 72: 965-976. Link: https://goo.gl/wVv3o8
  122. Huber TB, Hartleben B, Kim J, Schmidts M, Schermer B, et al. (2003) Nephrin and CD2AP associate with phosphoinositide 3-OH kinase and stimulate AKT-dependent signaling. Mol Cell Biol 23: 4917-4928. Link: https://goo.gl/SvpQ5f
  123. Furukawa-Hibi Y, Kobayashi Y, Chen C, Motoyama N (2005) FOXO transcription factors in cell-cycle regulation and the response to oxidative stress. Antioxid Redox Signal 7: 752-760. Link: https://goo.gl/i7zxwT
  124. Kops GJ, Burgering BM (1999) Forkhead transcription factors: new insights into protein kinase B (c-akt) signaling. J Mol Med 77: 656-665. Link: https://goo.gl/3OvTDC
  125. Accili D, Arden KC (2004) FoxOs at the crossroads of cellular metabolism, differentiation, and transformation. Cell 117: 421-426. Link: https://goo.gl/5SZqjL
  126. Brunet A, Sweeney LB, Sturgill JF, Chua KF, Greer PL, et al. (2004) Stress-dependent regulation of FOXO transcription factors by the SIRT1 deacetylase. Science 303: 2011-2015. Link: https://goo.gl/jk3x8w
  127. Grabiec AM, Angiolilli C, Hartkamp LM, van Baarsen LG, Tak PP, et al. (2015) JNK-dependent downregulation of FoxO1 is required to promote the survival of fibroblast-like synoviocytes in rheumatoid arthritis. Ann Rheum Dis 74: 1763-1771. Link: https://goo.gl/zP93qw
  128. Thornalley PJ (1998) Cell activation by glycated proteins. AGE receptors, receptor recognition factors and functional classification of AGEs. Cell Mol Biol 44: 1013-1023. Link: https://goo.gl/PbBhia
  129. Pricci F, Leto G, Amadio L, Iacobini C, Romeo G, et al. (2000) Role of galectin-3 as a receptor for advanced glycosylation end products. Kidney Int 77: S31-S39. Link: https://goo.gl/eQMoUO
  130. Schmidt AM, Vianna M, Gerlach M, Brett J, Ryan J, et al. (1992) Isolation and characterization of two binding proteins for advanced glycosylation end products from bovine lung which are present on the endothelial cell surface. J Biol Chem 267: 14987-14997. Link: https://goo.gl/3AuKaA
  131. Araki N, Higashi T, Mori T, Shibayama R, Kawabe Y, et al. (1995) Macrophage scavenger receptor mediates the endocytic uptake and degradation of advanced glycation end products of the Maillard reaction. Eur J Biochem 230: 408-415. Link: https://goo.gl/u7lp2D
  132. Miyazaki A, Nakayama H, Horiuchi S (2002) Scavenger receptors that recognize advanced glycation end products. Trends Cardiovasc Med 12: 258-262. Link: https://goo.gl/WGo46S
  133. Bucciarelli LG, Wendt T, Rong L, Lalla E, Hofmann MA, et al. (2002) RAGE is a multiligand receptor of the immunoglobulin superfamily: implications for homeostasis and chronic disease. Cell Mol Life Sci 59: 1117-1128. Link: https://goo.gl/CV3yfG
  134. Yoonhee Kim, Chaeyoung Kim, Sung Min Son, Hyundong Song, Hyun Seok Hong (2016) The novel RAGE interactor PRAK is associated with autophagy signaling in Alzheimer’s disease pathogenesis. Mol Neurodegener 11: 4. Link: https://goo.gl/tz68JP
  135. Taguchi A, Blood DC, del Toro G, Canet A, Lee DC, et al. (2000) Blockade of RAGE-amphoterin signalling suppresses tumour growth and metastases. Nature 405: 354-360. Link: https://goo.gl/t2vqr0
  136. Brett J, Schmidt AM, Yan SD, Zou YS, Weidman E, et al. (1993) Survey of the distribution of a newly characterized receptor for advanced glycation end products in tissues. Am J Pathol 143: 1699-1712. Link: https://goo.gl/mZpcN0
  137. Tóbon-Velasco JC, Cuevas E, Torres-Ramos MA (2014) Receptor for AGEs (RAGE) as mediator of NF-kB pathway activation in neuroinflammation and oxidative stress. CNS Neurol Disord Drug Targets 13: 1615-1626. Link: https://goo.gl/Fb7KgD
  138. Sugaya K, Fukagawa T, Matsumoto K, Mita K, Takahashi E,et al. (1994) Three genes in the human MHC class III region near the junction with the class II gene for receptor of advanced glycosylation end products, PBX2 homeobox gene and a notch homolog, human counterpart of mouse mammary tumor gene int-3. Genomics 23: 408-419. Link: https://goo.gl/mMVtns
  139. Srikrishna G, Huttunen HJ, Johansson L, Weigle B, Yamaguchi Y, et al. (2001) N-Glycans on the receptor for advanced glycation end products influence amphoterin binding and neurite outgrowth. J Neurochem 80: 998-1008. Link: https://goo.gl/kR9XTY
  140. Hudson BI, Kalea AZ, Del Mar Arriero M, Harja E, Boulanger E, et al. (2008) Interaction of the RAGE cytoplasmic domain with diaphanous-1 is required for ligand-stimulated cellular migration through activation of Rac1 and Cdc42. J Biol Chem 283: 34457-34468. Link: https://goo.gl/LZ5cDM
  141. Tang X, Qin Q, Xie X, He P (2015) Protective effect of sRAGE on fetal development in pregnant rats with gestational diabetes mellitus. Cell Biochem Biophys 71: 549-556. Link: https://goo.gl/pwHe0A
  142. Jin Q1, Chen H, Luo A, Ding F, Liu Z (2016) Correction: S100A14 Stimulates Cell Proliferation and Induces Cell Apoptosis at Different Concentrations via Receptor for Advanced Glycation End Products (RAGE). PLoS One 11: e0147881. Link: https://goo.gl/HpS8uv
  143. Zurolo E, Iyer A, Maroso M, Carbonell C, Anink JJ, et al. (2011) Activation of Toll-like receptor, RAGE and HMGB1 signalling in malformations of cortical development. Brain 134: 1015-1032. Link: https://goo.gl/c6z0K6
  144. Bierhaus A, Humpert PM, Morcos M, Wendt T, Chavakis T, et al. (2005) Understanding RAGE, the receptor for advanced glycation end products. J Mol Med (Berl) 83: 876-886. Link: https://goo.gl/PpE3kL
  145. Ramasamy R, Vannucci SJ, Yan SS, Herold K, Yan SF, et al. (2005) Advanced glycation end products and RAGE: a common thread in aging, diabetes, neurodegeneration, and inflammation. Glycobiology 15: 16R-28R. Link: https://goo.gl/3D6xST
  146. Collison KS, Parhar RS, Saleh SS, Meyer BF, Kwaasi AA, et al. (2002) RAGE-mediated neutrophil dysfunction is evoked by advanced glycation end products (AGEs). J Leukoc Biol 71: 433-444. Link: https://goo.gl/EnwV6B
  147. Tanji N, Markowitz GS, Fu C, Kislinger T, Taguchi A, et al. (2000) Expression of advanced glycation end products and their cellular receptor RAGE in diabetic nephropathy and nondiabetic renal disease. J Am Soc Nephrol 11: 1656-1666. Link: https://goo.gl/UdmqO1
  148. Weijing Cai, Qiao-Di Gao, Li Zhu, Melpomeni Peppa, Cijiang He, et al. (2002) Oxidative stress-inducing carbonyl compounds from common foods: novel mediators of cellular dysfunction. Mol Med 8: 337-346. Link: https://goo.gl/BtYr9f
  149. Loeser RF, Yammani RR, Carlson CS, Chen H, Cole A, et al. (2005) Articular chondrocytes express the receptor for advanced glycation end products: potential role in osteoarthritis. Arthritis Rheum 52: 2376-2385. Link: https://goo.gl/tJaszn
  150. Lin L, Park S, Lakatta EG (2009) RAGE signaling in inflammation and arterial aging. Front Biosci 14: 1403-1413. Link: https://goo.gl/2D5dpu
  151. Piperi C, Adamopoulos C, Dalagiorgou G, Diamanti-Kandarakis E, Papavassiliou AG (2012) Crosstalk between advanced glycation and endoplasmic reticulum stress: emerging therapeutic targeting for metabolic diseases. J Clin Endocrinol Metab 97: 2231–42. Link: https://goo.gl/PGGJ3A
  152. Li J, Schmidt AM (1997) Characterization and functional analysis of the promoter of RAGE, the receptor for advanced glycation end products. J Biol Chem 272: 16498-16506. Link: https://goo.gl/P9W9O1
  153. Ritthaler U, Deng Y, Zhang Y, Greten J, Abel M, et al. (1995) Expression of receptors for advanced glycation end products in peripheral occlusive vascular disease. Am J Pathol 146: 688-694. Link: https://goo.gl/XayyY4
  154. Kislinger T, Tanji N, Wendt T, Qu W, Lu Y, et al. (2001) Receptor for advanced glycation end products mediates inflammation and enhanced expression of tissue factor in vasculature of diabetic apolipoprotein E-null mice. Arterioscler Thromb Vasc Biol 21: 905-910. Link: https://goo.gl/8wy5EV
  155. Watson AM, Li J, Samijono D, Bierhaus A, Thomas MC, et al. (2014) Quinapril treatment abolishes diabetes-associated atherosclerosis in RAGE/apolipoprotein E double knockout mice. Atherosclerosis 235: 444-448. Link: https://goo.gl/m3C1LC
  156. Buschmann K, Tschada R, Metzger MS, Braach N, Kuss N, et al. (2014) RAGE controls leukocyte adhesion in preterm and term infants. BMC Immunol 15: 53. Link: https://goo.gl/Oj7ITB
  157. Orlova VV, Choi EY, Xie C, Chavakis E, Bierhaus A, et al. (2007) A novel pathway of HMGB1-mediated inflammatory cell recruitment that requires Mac-1-integrin. EMBO J 26: 1129-1139. Link: https://goo.gl/v54XzA
  158. Pullerits R, Brisslert M, Jonsson IM, Tarkowski A (2006) Soluble receptor for advanced glycation end products triggers a proinflammatory cytokine cascade via beta2 integrin Mac-1. Arthritis Rheum 54: 3898-3907. Link: https://goo.gl/exQYhX
  159. Bonaldi T, Talamo F, Scaffidi P, Ferrera D, Porto A, et al. (2003) Monocytic cells hyperacetylate chromatin protein HMGB1 to redirect it towards secretion. EMBO J 22: 5551-5560. Link: https://goo.gl/Nc3NNJ
  160. Carlos TM, Harlan JM (1994) Leukocyte-endothelial adhesion molecules. Blood 84: 2068-2101. Link: https://goo.gl/9tMb9X
  161. Ramasamy R, Yan SF, Herold K, Clynes R, Schmidt AM (2008) Receptor for Advanced Glycation End Products: Fundamental Roles in the Inflammatory Response: Winding the Way to the Pathogenesis of Endothelial Dysfunction and Atherosclerosis. Ann NY Acad Sci 1126: 7-13. Link: https://goo.gl/ecuWx3
  162. Figarola JL, Shanmugam N, Natarajan R, Rahbar S (2007) Anti-Inflammatory Effects of the Advanced Glycation End Product Inhibitor LR-90 in Human Monocytes. Diabetes 56: 647-655. Link: https://goo.gl/D78HFr
  163. Lalla E, Lamster IB, Feit M, Huang L, Spessot A, et al. (2000) Blockade of RAGE suppresses periodontitis-associated bone loss in diabetic mice. J Clin Invest 105: 1117-1124. Link: https://goo.gl/hyui1B
  164. Wautier JL, Zoukourian C, Chappey O, Wautier MP, Guillausseau PJ, et al. (1996) Receptor-mediated endothelial cell dysfunction in diabetic vasculopathy: soluble receptor for advanced glycation end products blocks hyperpermeability in diabetic rats. J Clin Invest 97: 238-243. Link: https://goo.gl/efguat  
  165. Wendt T, Harja E, Bucciarelli L, Qu W, Lu Y, et al. (2006) Rage modulates vascular inflammation and atherosclerosis in a murine model of type 2 diabetes. Atherosclerosis 185: 70-77. Link: https://goo.gl/AhqCx
  166. J
  167. Chavakis T, Bierhaus A, Nawroth PP (2004) RAGE (receptor for advanced glycation end products): a central player in the inflammatory response. Microbes Infect 6: 1219-1225 Link: https://goo.gl/rkbvA5
© 2017 Guo H, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
 

Help ?