Next Article in Journal
Genetic Approaches Using Zebrafish to Study the Microbiota–Gut–Brain Axis in Neurological Disorders
Next Article in Special Issue
Inhibition of Cochlear HMGB1 Expression Attenuates Oxidative Stress and Inflammation in an Experimental Murine Model of Noise-Induced Hearing Loss
Previous Article in Journal
Neutrophil Adhesion and the Release of the Free Amino Acid Hydroxylysine
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Immune Tolerance Role of the HMGB1-RAGE Axis

1
Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030, USA
2
Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
*
Author to whom correspondence should be addressed.
Cells 2021, 10(3), 564; https://doi.org/10.3390/cells10030564
Submission received: 5 February 2021 / Revised: 26 February 2021 / Accepted: 3 March 2021 / Published: 5 March 2021
(This article belongs to the Special Issue Regulation of HMGB1 Release in Health and Diseases)

Abstract

:
The disruption of the immune tolerance induces autoimmunity such as systemic lupus erythematosus and vasculitis. A chromatin-binding non-histone protein, high mobility group box 1 (HMGB1), is released from the nucleus to the extracellular milieu in particular environments such as autoimmunity, sepsis and hypoxia. Extracellular HMGB1 engages pattern recognition receptors, including Toll-like receptors (TLRs) and the receptor for advanced glycation endproducts (RAGE). While the HMGB1-RAGE axis drives inflammation in various diseases, recent studies also focus on the anti-inflammatory effects of HMGB1 and RAGE. This review discusses current perspectives on HMGB1 and RAGE’s roles in controlling inflammation and immune tolerance. We also suggest how RAGE heterodimers responding microenvironments functions in immune responses.

1. Immune Tolerance and Autoimmunity

Failure of immune tolerance results in lymphocyte reactions against self-antigens called autoimmunity and the diseases caused by autoimmunity are referred to as autoimmune diseases. Autoimmune reactions are triggered by environmental factors, such as infections, in genetically susceptible individuals. Autoimmune diseases are classified as a systemic or organ-specific disease, depending on the distribution of the autoantigens that are recognized [1]. For example, autoantibodies against ubiquitous antigens, including nuclear components typically cause systemic diseases, such as systemic lupus erythematosus (SLE) and vasculitis. Autoantibodies or T cell responses against self-antigens with restricted tissue distribution lead to organ-specific diseases, such as autoimmune thyroiditis, type 1 diabetes, and myasthenia gravis. The adaptive immune response including autoreactive T lymphocytes, circulating autoantibodies and immune complexes is generally thought to be responsible for tissue injury in autoimmune diseases.
Most autoimmune diseases are polygenic, and numerous susceptibility genes contribute to the predisposition to disease development. SLE is a systemic disease in which autoantibodies such as anti-DNA antibodies form immune complexes and is characterized by its heterogeneous clinical manifestations, including cutaneous, kidney, central nervous system [2,3,4]. Though the pathogenesis of SLE has not been fully elucidated, there is abnormal clearance of apoptotic debris which induce anti-DNA antibody production and lead to inflammation causing clinical symptoms [5]. It frequently occurs in young women, and various immunosuppressive drugs are used for treatment [6]. The impact of genetic susceptibility on the development of SLE has been well demonstrated in a number of large-scale genome-wide association studies (GWAS) [7]. Notably, a functional SNP located in the 3′ untranslated region of TLR7 was associated with SLE. The Interferon regulatory factor 5 (IRF5) genetic locus carries multiple functional polymorphisms that potentially associate with SLE [8]. Genetic deficiency of C1q predisposes strongly to SLE and C1q polymorphisms are associated with more severe SLE, low serum C1q, and low levels of total hemolytic complement [9].
Antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) is another systemic autoimmune disease. AAV is characterized by ANCA production and small- and medium-sized blood vessel inflammation [10,11]. AAV commonly causes life-threatening kidney failure or pulmonary hemorrhage and affects short- and long-term mortality. GWAS has revealed that HLA, α1-antitrypsin and proteinase 3 are associated with ANCA specificity [12].
Several tolerance mechanisms have been studied in monocytes/macrophages contributing to scavenging, inflammation, and anti-pathogen defenses [13,14,15]. Ineffectual clearance of immune complexes and accumulation of apoptotic cells expose the immune system to various autoantigens [7]. The repeated or chronic activation of Toll-like receptors (TLRs) by a bacterial product such as lipopolysaccharide (LPS) induce immune tolerance to the secondary infection [16,17,18]. These mechanisms are important to prevent prolonged or repeated activation of TLRs leading to uncontrolled inflammation and subsequent damages.
Macrophage polarization is another way which maintains immune homeostasis. Monocytes can be polarized through different activation programs to the classical pro-inflammatory M1 macrophages and the anti-inflammatory M2 macrophages [19,20]. Pro-inflammatory macrophages contribute to immune protection in infection, but also to disease pathogenesis in autoimmune diseases, atherosclerosis and Alzheimer’s disease and anti-inflammatory macrophages are important for cessation of inflammation and tissue repair.
High mobility group box 1 (HMGB1) is a damage-associated molecular pattern (DAMPs) and high in autoimmune diseases, tissue injury and infection [2,21,22]. HMGB1 exhibits many other functions in immune response, either pro-inflammatory or anti-inflammatory. In this review, we describe the role of HMGB1 as a positive and negative regulator for inflammatory autoimmune diseases. We describe the tolerance mechanism of HMGB1 that provides potential therapeutics for autoimmune diseases.

2. HMGB1

HMGB1 is an evolutionary conserved nuclear protein that binds to DNA to maintain chromatin structure, involves DNA repair, and indirectly regulates the activities of various transcription factors such as NF-κB and glucocorticoid receptors [23,24]. HMGB1 is highly conserved in mammals [25]. HMGB1 has 215 amino acid residues and forms two DNA binding domains (A box [9–79 amino acid], B box [95–163 amino acid]) and a C-terminal acidic tail (186–215 amino acid) [26]. HMGB1 is released passively from necrotic cells or actively from activated dendritic cells and macrophages, then relates to various pathological conditions [27]. Caspase 3/7-mediated programmed cell death, autophagy, pyroptosis cause the passive release of HMGB1. Active secretion of HMGB1 occurs through the extensive post-translational modifications (PTM) such as acetylation, phosphorylation, methylation and oxidation that determine the localization of HMGB1. PTM-mediated active secretion of HMGB1 is mediated through secretory lysosomes [28]. The oxidation status of HMGB1 and HMGB1 secretion kinetics also influences its immune function [28]. Although the exact active secretion mechanism of HMGB1 remains elusive, a recent study has revealed that C5a engagement with its receptor C5aR2 in macrophages upon infection induces upregulation of HMGB1 expression and release through intracellular signaling [29] (Figure 1).
Small molecules can inhibit HMGB1 secretion [28]. Glycyrrhizin inhibits the cytoplasmic transduction of HMGB1 and suppresses the expression of inflammatory cytokines [28]. Anti-inflammatory drug, Metformin binds HMGB1 and inhibits nuclear HMGB1 translocation to the cytosol [30,31]. The nuclear factor-erythroid 2-related factor 2 (Nrf2)/hemeoxygenase-1 (HO-1) pathway also plays an important role in the HMGB1 secretion. HO-1 suppresses the translocation and secretion of HMGB1 [32,33]. Nrf2 is a redox-sensitive transcription factor for HO-1 [34,35] (Figure 1). Anti-inflammatory vitamin D (1,25-dihydroxy vitamin D) inhibits LPS-induced HMGB1 secretion in macrophages through the Nrf2/HO-1 pathway [34].
Extracellular HMGB1 can bind with the receptor for advanced glycation endproducts (RAGE), TLRs, and cytosolic DNA/RNA sensors mediating inflammation [36,37]. Disulfide HMGB1 binds to MD2 in the TLR4 receptor complex and induces cytokine production [38]. All HMGB1 redox forms bind to RAGE and HMGB1-RAGE interaction results in uptake in endosomes that present TLR7 and 9 [39]. Without any doubt, HMGB1 is a crucial mediator for the innate immune system and an attractive target for therapy in many disease states, including sepsis, ischemia, arthritis, autoimmune diseases, neurodegenerative diseases, metabolic disorders and cancer [40]. Gain- and loss-of-function analysis showed that HMGB1 exacerbated the severity of renal disease and autoimmunity in the murine model of SLE [41]. Extracellular blockades such as neutralizing mouse/rat/humanized anti-HMGB1 antibodies, receptor blocking HMGB1 A box and FSSE tetramer have the distinct potential to improve clinical outcome in multiple inflammatory diseases [38,42,43,44].
Through extensive studies by others, extracellular HMGB1 is highly inclined to bind many molecules, and most other receptors are presented in a complex form with HMGB1. Besides, RAGE has multi ligands. Current studies suggest that the alternative mechanistic explanation for HMGB1 and RAGE axis is that this interaction provides both pro-inflammatory and anti-inflammatory functions—the critical functional difference may be caused by HMGB1-RAGE attached to partner molecules.

3. RAGE Functions Following HMGB1 Engagement

RAGE is an approximately 40 kDa of a pattern recognition receptor belonging to the immunoglobulin superfamily [45]. RAGE is evolutionarily present only in mammals [46], and its deficiency in mice show normal development [47,48]. RAGE is a type I transmembrane protein composed of three extracellular immunoglobulin-like domains (V, C1, and C2), a single transmembrane helix, and a C-terminal short domain, and exists in lipid rafts [45,49] (Figure 2). RAGE has various ligands such as HMGB1, AGEs, S100/calgranulin family, Mac-1, β sheet fibrils, and LPS can bind predominantly with the V domain of RAGE [50,51]. Upon ligand binding, RAGE activates multiple intracellular signaling pathways involving in the small guanine nucleotide triphosphatases (GTPases) ras-related C3 botulinum toxin substrate 1 (Rac1)/cell division control protein 42 (Cdc42), Ras-mediated extracellular signal-regulated kinase 1/2 (ERK1/2), Phosphoinositide 3-kinase (PI3-K)/Akt, stress-activated protein kinase/c-Jun-NH2-terminal kinase (SAPK/JNK), p38 mitogen-activated protein kinase (MAPK), NF-κB and caspases [52]. The RAGE cytoplasmic domain regulates cell signaling and function through binding with adaptor proteins including diaphanous homolog 1 (Diaph1), toll-interleukin 1 receptor domain containing adaptor protein (TIRAP), and myeloid differentiation primary response 88 (MyD88) [51,53]. RAGE is a constitutive multimer on the plasma membrane [54], and such multimers comprise at least four RAGE molecules before ligand binding [51,55]. It seems that the multimerization of RAGE could make ligand multimers possible to bind and sustain downstream signal transduction [51,52]. RAGE also can form heterodimers with other membrane proteins and exert various biological functions. It is reported that DNAX-activating protein 10, a transmembrane protein, also bound to RAGE resulted in an enhancement of Akt activation while homomultimeric RAGE led to the activation of caspase-8 [56] (Figure 3).
RAGE itself contains a tolerogenic system to control overreactions. RAGE has three variants: full length, N-terminally truncated, and C-terminally truncated. Endogenously secreted RAGE, a spliced variant of RAGE, is the C-terminally truncated form of RAGE secreted from the cell and has a V-domain essential for binding ligands [57]. Another isoform, soluble RAGE (sRAGE), is cleaved from cell-surface RAGE by matrix metalloproteinases [58]. Secreted RAGE and truncated RAGE serve as a decoy receptor sequestering ligands and inhibiting signal transduction [57,58,59] (Table 1).
In 1995, Hori et al. first reported that HMGB1 could bind to RAGE [60]. Amino acids 150–183 of HMGB1 are responsible for binding to RAGE for invasive migration and growth of tumor cells through the activation of p38 MAPK and Erk1/2 [61,62]. HMGB1 induced phosphorylation of endogenous RAGE cytosolic domain at Ser391 by PKCζ, and seemed to interact with TIRAP and MyD88, then transduce signals to the downstream molecules such as NF-κB [63]. RAGE is also localized in mitochondria of tumor cells [64], and interaction between HMGB1 and RAGE regulates cellular metabolism and promotes tumor growth by enhancing ATP production. The interaction HMGB1 and RAGE also has been reported to trigger neutrophil-mediated injury amplification following necrosis [65].
Therapeutics targeting RAGE has been developed. Blockade of RAGE signaling by antibody or soluble RAGE also has been shown to the efficacy in several disease models including diabetic complications, sepsis, and autoimmunity [66,67,68,69]. Clinical development may have progressed most in small-molecule inhibitors of RAGE. TTP488, which was discovered by the pharmaceutical industry, is an orally active antagonist of RAGE-RAGE ligand interaction [70] and tested in Phase 2 clinical trials to treat Alzheimer’s disease [71]. FPS-ZM1 was found by screening for molecules inhibiting amyloid β binding to the V domain of RAGE [72]. The efficacy of FPS-ZM1 for cerebral hemorrhage, cerebral ischemia, emphysema, cancer, and inflammatory bowel diseases, have been reported [73,74,75,76,77]. Another molecule inhibiting RAGE signaling is a DNA oligonucleotide aptamer (RAGE-aptamer) [78]. RAGE-aptamer has been reported to reverse and prevent the development of diabetic nephropathy in vivo, and suppressed the AGE-induced reactive oxygen species (ROS) generation and inflammatory and fibrotic reactions in vitro [78]. RAGE-aptamer also attenuates melanoma growth and liver metastasis in vivo by reducing tumor-associated angiogenesis and macrophage infiltration by suppressing the AGE-RAGE system [79].

4. The HMGB1-RAGE Axis in SLE

RAGE provides a functional platform for crosstalk with other HMGB1 receptors that exist in organelles. The HMGB1-DNA complexes binding with RAGE on the cell surface result in internalizing into the cytosol and interacting with TLR9, which exist in the endosome, augment type 1 IFN production through a mechanism dependent on MyD88 [80]. Type 1 IFN plays an important role in the pathogenesis of SLE. The DNA-containing immune complexes are also a key factor activation of autoreactive B cells and the induction of type I IFN dependent on RAGE engagement [80]. Further, HMGB1-DNA internalization by RAGE also has been reported in inflammatory monocytes exposed to serum from patients with SLE [81]. Thus, HMGB1 and RAGE are involved in autoimmunity by transmitting intracellular signals and acting as a carrier between the extracellular and intracellular compartments.
Lupus nephritis (LN), which is characterized by renal deposition of immune complexes, is a refractory complication of SLE, which causes end-stage renal disease resulting in lower survival rates and quality of life [6]. It has been reported that anti-DNA antibody binding with HMGB1 exhibited a synergistic proinflammatory effect on mesangial cells of LN patients in a RAGE dependent manner [82]. They also found enhanced susceptibility of lupus prone MRL/lpr mice as compared to normal mice derived mesangial cells to anti-DNA antibody and LPS stimulation, in addition to significantly increased expression of TLR4 [82]. Increased HMGB1 expression deteriorated the severity of SLE via enhancing macrophage inflammatory response, and RAGE played a critical role in HMGB1-mediated macrophage inflammatory response [41]. Treatment with sRAGE, the soluble extracellular domains of RAGE, which blocks ligands interaction with RAGE demonstrated significant improvement of nephritis in (NZB/NZW) F1 lupus-prone mice [69]. They have shown that sRAGE interrupted the nuclear translocation of NF-κB in the kidney, resulting in a reduction in the expression of downstream genes of NF-κB in vivo and in vitro. Moreover, plasma sRAGE level in patients with SLE was significantly lower than those in healthy controls and negatively correlated with SLE disease activity, suggesting a rationale for sRAGE as a therapeutic [83,84]. Though blocking RAGE signaling seems a promising approach to treat LN, there is a controversial report. The RAGE knock-out mice with C57BL/6 background lpr mice exacerbated of autoantibody titers and nephritis was observed [85]. RAGE knock-out lupus mice exhibited a delay in apoptosis of CD3 + B220 + CD4 − CD8 − autoreactive T cells, and an increase in these pathogenic T cells was thought to exacerbate the disease [85].

5. The HMGB1-RAGE Axis in Autoimmune Vasculitis

HMGB1 plays an important role in the pathogenic mechanism of autoimmune vasculitis. Like lupus, circulating HMGB1 levels have been reported to be increased and closely associated with the disease activity of AAV [86,87]. HMGB1 could prime neutrophils by increasing ANCA antigens translocation to the cell surface. The primed neutrophils could be further induced by ANCA, resulting in the respiratory burst and degranulation in TLR4- and RAGE-dependent manner through the MyD88/NF-κB pathway [88]. Recently, the abnormal regulation of neutrophil extracellular traps (NETs), generated by ANCA-activated neutrophils, have been recognized to contribute to the pathogenesis of AAV [89,90]. NETs can stick to the endothelium and cause tissue damage during inflammation [91]. It is also reported that NETs are associated with thrombosis in AAV patients as histones and DNA within NETs can bind platelets and blood coagulants [90,92]. HMGB1 exerts effects on NETs formation through interaction with TLR2, TLR4, and RAGE, and the process is NADPH oxidase dependent [93]. Another study suggests that HMGB1 from activated platelets commit neutrophils to NET generation through RAGE, resulting in preventing depletion of mitochondrial potential, autophagosome formation, and prolonging neutrophil survival [94]. Thus, the HMGB1-RAGE axis can potentiate ANCA-inducing NETs formation and may be involved in the pathogenesis of AAV.
Peschel and colleagues have revealed the high prevalence of autoantibodies to lysosome-associated membrane protein-2 (LAMP-2) in ANCA-negative pauci-immune focal necrotizing glomerulonephritis [95]. LAMP-2 is a heavily glycosylated membrane protein that traffics from the cell surface to lysosomes, where it is critical for cellular homeostasis and responses to stress by participating in autophagy [96]. LAMP-2 is an endocytic receptor on human monocyte-derived dendritic cells that routes cargo into immunogenic exosomes while reducing surface expression of antigen-derived peptides [97]. A perspective from immune tolerance of the HMGB1-RAGE axis might provide a novel understanding of the pathogenesis of AAV. There is an association of the HMGB1-RAGE axis with autophagy [98,99].
The anti-inflammatory effects of HMGB1 blockades, including anti-HMGB1 monoclonal antibody and glycyrrhizin, have been shown in a mouse model of cutaneous vasculitis [100].

6. The HMGB1-RAGE Axis in Ischemic Diseases

The HMGB1-RAGE axis also involves in the pathophysiology of ischemic diseases. Watanabe et al. first described the relationship between HMGB1 and ischemia-reperfusion (I/R) injury [101]. During hepatic I/R injury, HMGB1 increased and translocated from nuclear to the cytoplasm as early as one hour after ischemia [102]. Inhibition of HMGB1 activity with neutralizing antibody decreased liver damage. In contrast, the administration of recombinant HMGB1 worsened I/R injury [102]. Blood HMGB1 level is also elevated in patients with cerebral or myocardial ischemia. Kim et al. reported the relationship between HMGB1 and brain inflammatory injury in the middle cerebral artery occlusion/reperfusion-induced injury model [103]. HMGB1 also promotes angiogenesis and neurovascular remodeling via endothelial progenitor cells by binding to the RAGE [104].
The HMGB1-RAGE axis also could take part in hypoxia-induced organ damages. Hypoxia induces HMGB1 and AGE accumulation which further formed a complex with RAGE and activates several downstream pathways including NF-κB, hypoxia-inducible factor-1 (HIF-1α), ERK1/2, and Akt signaling [105,106]. It has been demonstrated that the RAGE promoter region contains at least one functional HIF-1 binding site, and HIF-1α down-regulation drastically decreased RAGE induction by hypoxia in neurons, suggesting that hypoxic stimulation of RAGE expression could be mediated by Hif1α [107]. The renal hypoxia promotes ROS formation, and it appears that oxidative stress is a central regulator of HMGB1′s translocation, release, and activity in inflammation and cell death [35]. RAGE-dependent vascular perturbation in hypoxia has also been identified, and the vascular dysfunction may amplify hypoxic HMGB1-RAGE mediated organ damages [108]. Thus, this axis may serve as a master regulator of inflammatory stress triggered by hypoxia.
Dobutamine mediates HO-1 induction via Nrf-2 translocation to inhibit the HMGB1 release in rat myocardial I/R injury. These suggest that Nrf-2-HO-1 axis may serve as a regulator for the HMGB1-RAGE axis and provide a potential therapeutic target in ischemic diseases [109,110].

7. Tolerogenic Role of the HMGB1-RAGE Axis

Recent studies demonstrate that several molecules neutralize extracellular HMGB1 or convert its pro-inflammatory functions to anti-inflammatory functions. While the HMGB1-RAGE axis drives inflammation, it also could regulate the immune tolerance by inducing anti-inflammatory macrophages. RAGE could exert various functions by interacting with other membrane receptors and the HMGB1-RAGE axis exerts tolerogenic functions in certain conditions. Under particular microenvironments or with proximal receptors, RAGE can induce immune tolerance. G-protein-coupled receptors involved in the cAMP signal pathway and the phosphatidylinositol signal pathway, interact with RAGE. Mainly, formyl-peptide-receptors play an essential role in amyloid β-induced ERK1/2 phosphorylation and changes in cAMP levels in glial cells by interacting with RAGE [111]. Leukotriene B4 (LTB4) receptor 1 (BLT1) interacts with RAGE and induces proinflammatory cytokines and chemokines in vitro/ex vivo [112]. Both LTB4-dependent ERK1/2 phosphorylation in neutrophils and LTB4-dependent neutrophil accumulation in a murine peritonitis model were attenuated in RAGE-deficient mice compared with wild-type mice [112] (Figure 3). Moreover, it has been reported the paradoxical role of HMGB1 in the tumor microenvironment [113]. HMGB1 contributes to the protumoral activities of the M2 macrophage phenotype by a RAGE-dependent mechanism [114]. Released HMGB1 due to hypoxia promotes M2-like macrophage accumulation and an IL-10 rich milieu by selectively signaling through RAGE [106]. We have recently demonstrated that complement component C1q can form a multimolecular signaling complex with HMGB1, RAGE, and Leukocyte-Associated Ig-like Receptor-1 (LAIR-1) in lipid rafts, and suppress inflammation by promoting M2-like macrophage polarization [115] (Table 1). We have also shown that HMGB1 promotes leukotriene production, induces IRF5 in a RAGE-dependent manner, while it produces specialized pro-resolving lipid mediators (SPMs), which have a negative effect on leukotriene synthesis and help in the resolution of inflammation [116,117,118] (Figure 3). These observations indicate that RAGE could not only facilitate inflammation but also resolve inflammation. Thus, the immune homeostasis maintaining the role of RAGE in diverse microenvironments should be examined in future studies. Though further studies are required to elucidate the environmental factors that determine the various function of the HMGB1-RAGE axis, C1q-LAIR-1 is one of the most important partners.
The HMGB1-RAGE axis has a role in regulatory T cells (Tregs), which is a subset of CD4+ T cells and dampening T-cell immune responses against self-antigens and maintaining immunological tolerance. Wild et al. have found that Tregs express significantly higher RAGE levels on the cell surface than conventional T cells, and HMGB1 induces Tregs migration and prolonged their survival in a RAGE-dependent manner [119]. Yang et al. reported that CD163 is an anti-inflammatory receptor for HMGB1-haptoglobin complexes [120]. Haptoglobin, the acute phase protein that binds extracellular hemoglobin and targets cellular uptake through CD163, also binds HMGB1. Haptoglobin is an endogenous HMGB1 binding protein that directs HMGB1 to a CD163-dependent receptor pathway that elicits HO-1 and IL-10 in the monocyte-macrophage lineage [120]. Another study has shown that binding of HMGB1 by soluble CD52, a glycophosphatidylinositol-anchored glycoprotein, promotes ligation of soluble CD52 with the sialic acid-binding Ig-like lectin-10 receptor and suppression of T cell function [121] (Table 1).
Table 1. Immune tolerance functions of the HMGB1-RAGE axis.
Table 1. Immune tolerance functions of the HMGB1-RAGE axis.
MoleculeMode of ActionReference
Soluble RAGEDecoy receptor for RAGE[59]
C1qInduce anti-inflammatory macrophage polarization[115,118]
HaptoglobinBind with CD163, activates heme oxygenase-1 IL-10 productions[120]
Soluble CD52Engage with the sialic acid-binding Ig-like lectin-10 receptor and suppress T cell function[121]
High mobility group box 1, HMGB1; receptor for advanced glycation endproducts, RAGE.
HMGB1 also demonstrates immunosuppressive activities through myeloid-derived suppressor cells (MDSCs) present in patients with solid tumors and contributes to immune suppression [122]. In murine tumor systems, HMGB1 foments the development of MDSC from bone marrow progenitor cells, enhances crosstalk between MDSC and macrophages by increasing MDSC production of IL-10, and reduces the expression of L-selectin on circulating T cells [123].

8. Conclusions

HMGB1 is the best well-characterized DAMPs for more than three decades. The unique feature of HMGB1 may operate in the opposite direction as an alarm signal for the environment. Particular receptors, ligands that are evolutionally closed with HMGB1 may maintain homeostasis as well as immune tolerance. For translating to therapeutics targeting the HMGB1-RAGE axis, not only simple inhibition but other strategy focusing on RAGE partners or individual intracellular molecules might be required due to the diversity of this axis.

Author Contributions

Conceptualization, writing, visualization, H.W. and M.S.; organization, supervision, M.S.; funding acquisition, M.S. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grant from the National Institute of Allergy and Infectious Diseases of the National Institutes of Health [R01AI135063].

Acknowledgments

The authors grateful to Betty Diamond for her comments and editing of this manuscript. Schematic figures were created with Biorender (© BioRender—biorender.com, accessed on 26 February 2021).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Abbas, A.K.; Lichtman, A.H.; Pillai, S.; Baker, D.L.; Baker, A. Cellular and Molecular Immunology, 9th ed.; Elsevier: Philadelphia, PA, USA, 2018; pp. 325–348. [Google Scholar]
  2. Liu, T.; Son, M.; Diamond, B. HMGB1 in Systemic Lupus Erythematosus. Front. Immunol. 2020, 11, 1057. [Google Scholar] [CrossRef]
  3. DeGiorgio, L.A.; Konstantinov, K.N.; Lee, S.C.; Hardin, J.A.; Volpe, B.T.; Diamond, B. A subset of lupus anti-DNA antibodies cross-reacts with the NR2 glutamate receptor in systemic lupus erythematosus. Nat. Med. 2001, 7, 1189–1193. [Google Scholar] [CrossRef]
  4. Kim, S.J.; Lee, K.; Diamond, B. Follicular Helper T Cells in Systemic Lupus Erythematosus. Front. Immunol. 2018, 9, 1793. [Google Scholar] [CrossRef] [Green Version]
  5. Baumann, I.; Kolowos, W.; Voll, R.E.; Manger, B.; Gaipl, U.; Neuhuber, W.L.; Kirchner, T.; Kalden, J.R.; Herrmann, M. Impaired uptake of apoptotic cells into tingible body macrophages in germinal centers of patients with systemic lupus erythematosus. Arthritis Rheum. 2002, 46, 191–201. [Google Scholar] [CrossRef]
  6. Mok, C.C. Towards new avenues in the management of lupus glomerulonephritis. Nat. Rev. Rheum. 2016, 12, 221–234. [Google Scholar] [CrossRef]
  7. Kwon, Y.C.; Chun, S.; Kim, K.; Mak, A. Update on the Genetics of Systemic Lupus Erythematosus: Genome-Wide Association Studies and Beyond. Cells 2019, 8, 1180. [Google Scholar] [CrossRef] [Green Version]
  8. Kim, S.J. SLE-associated risk factors affect DC function. Curr. Rheumatol. Rep. 2019, 21, 4. [Google Scholar] [CrossRef]
  9. Son, M.; Kim, S.J.; Diamond, B. SLE-associated risk factors affect DC function. Immunol. Rev. 2016, 269, 100–117. [Google Scholar] [CrossRef]
  10. Ball, G.V.; Fessler, B.J.; Bridges, S.L. Oxford Textbook of Vasculitis, 3rd ed.; Oxford University Press: Oxford, UK, 2014; pp. 61–69. [Google Scholar]
  11. Jennette, J.; Falk, R.; Bacon, P.; Basu, N.; Cid, M.; Ferrario, F.; Flores-Suarez, L.; Gross, W.; Guillevin, L.; Hagen, E.; et al. 2012 Revised International Chapel Hill Consensus Conference Nomenclature of Vasculitides. Arthritis Rheum. 2013, 65, 1–11. [Google Scholar] [CrossRef]
  12. Lyons, P.A.; Rayner, T.F.; Trivedi, S.; Holle, J.U.; Watts, R.A.; Jayne, D.R.; Baslund, B.; Brenchley, P.; Bruchfeld, A.; Chaudhry, A.N.; et al. Genetically distinct subsets within ANCA-associated vasculitis. N. Engl. J. Med. 2012, 367, 214–223. [Google Scholar] [CrossRef] [Green Version]
  13. Van Furth, R.; Cohn, Z.A. The origin and kinetics of mononuclear phagocytes. J. Exp. Med. 1968, 128, 415–435. [Google Scholar] [CrossRef]
  14. Medvedev, A.E.; Kopydlowski, K.M.; Vogel, S.N. Inhibition of lipopolysaccharide-induced signal transduction in endotoxin-tolerized mouse macrophages: Dysregulation of cytokine, chemokine, and toll-like receptor 2 and 4 gene expression. J. Immunol. 2000, 164, 5564–5574. [Google Scholar] [CrossRef] [Green Version]
  15. Kelly, A.; Gunaltay, S.; McEntee, C.P.; Shuttleworth, E.E.; Smedley, C.; Houston, S.A.; Fenton, T.M.; Levison, S.; Mann, E.R.; Travis, M.A. Human monocytes and macrophages regulate immune tolerance via integrin αvβ8-mediated TGFβ activation. J. Exp. Med. 2018, 215, 2725–2736. [Google Scholar] [CrossRef] [Green Version]
  16. Butcher, S.K.; O’Carroll, C.E.; Wells, C.A.; Carmody, R.J. Toll-Like Receptors Drive Specific Patterns of Tolerance and Training on Restimulation of Macrophages. Front. Immunol. 2018, 9, 933. [Google Scholar] [CrossRef] [Green Version]
  17. Sly, L.M.; Rauh, M.J.; Kalesnikoff, J.; Song, C.H.; Krystal, G. LPS-induced upregulation of SHIP is essential for endotoxin tolerance. Immunity 2004, 21, 227–239. [Google Scholar] [CrossRef] [Green Version]
  18. Dobrovolskaia, M.A.; Medvedev, A.E.; Thomas, K.E.; Cuesta, N.; Toshchakov, V.; Ren, T.; Cody, M.J.; Michalek, S.M.; Rice, N.R.; Vogel, S.N. Induction of in vitro reprogramming by Toll-like receptor (TLR)2 and TLR4 agonists in murine macrophages: Effects of TLR “homotolerance” versus “heterotolerance” on NF-kappa B signaling pathway components. J. Immunol. 2003, 170, 508–519. [Google Scholar] [CrossRef] [Green Version]
  19. Mills, C.D.; Kincaid, K.; Alt, J.M.; Heilman, M.J.; Hill, A.M. M-1/M-2 macrophages and the Th1/Th2 paradigm. J. Immunol. 2000, 164, 6166–6173. [Google Scholar] [CrossRef] [Green Version]
  20. Sica, A.; Mantovani, A. Macrophage plasticity and polarization: In vivo veritas. J. Clin. Invest. 2012, 122, 787–795. [Google Scholar] [CrossRef]
  21. Jiang, W.; Pisetsky, D.S. Expression of high mobility group protein 1 in the sera of patients and mice with systemic lupus erythematosus. Ann. Rheum. Dis. 2008, 67, 727–728. [Google Scholar] [CrossRef]
  22. Chen, T.; Fu, L.X.; Guo, Z.P.; Yin, B.; Cao, N.; Qin, S. Involvement of high mobility group box-1 in imiquimod-induced psoriasis-like mice model. J. Dermatol. 2017, 44, 573–581. [Google Scholar] [CrossRef]
  23. Urbonaviciute, V.; Voll, R.E. High-mobility group box 1 represents a potential marker of disease activity and novel therapeutic target in systemic lupus erythematosus. J. Intern. Med. 2011, 270, 309–318. [Google Scholar] [CrossRef]
  24. Harris, H.E.; Andersson, U.; Pisetsky, D.S. HMGB1: A multifunctional alarmin driving autoimmune and inflammatory disease. Nat. Rev. Rheumatol. 2012, 8, 195–202. [Google Scholar] [CrossRef]
  25. Tripathi, A.; Shrinet, K.; Kumar, A. HMGB1 protein as a novel target for cancer. Toxicol. Rep. 2019, 6, 253–261. [Google Scholar] [CrossRef]
  26. Bianchi, M.E.; Falciola, L.; Ferrari, S.; Lilley, D.M. The DNA binding site of HMG1 protein is composed of two similar segments (HMG boxes), both of which have counterparts in other eukaryotic regulatory proteins. EMBO J. 1992, 11, 1055–1063. [Google Scholar] [CrossRef] [PubMed]
  27. Wang, H.; Bloom, O.; Zhang, M.; Vishnubhakat, J.M.; Ombrellino, M.; Che, J.; Frazier, A.; Yang, H.; Ivanova, S.; Borovikova, L.; et al. HMG-1 as a late mediator of endotoxin lethality in mice. Science 1999, 285, 248–251. [Google Scholar] [CrossRef]
  28. Kwak, M.S.; Kim, H.S.; Lee, B.; Kim, Y.H.; Son, M.; Shin, J.S. Immunological Significance of HMGB1 Post-Translational Modification and Redox Biology. Front. Immunol. 2020, 11, 1189. [Google Scholar] [CrossRef]
  29. Zhang, T.; Wu, K.Y.; Ma, N.; Wei, L.L.; Garstka, M.; Zhou, W.; Li, K. The C5a/C5aR2 axis promotes renal inflammation and tissue damage. JCI Insight 2020, 5. [Google Scholar] [CrossRef] [Green Version]
  30. Horiuchi, T.; Sakata, N.; Narumi, Y.; Kimura, T.; Hayashi, T.; Nagano, K.; Liu, K.; Nishibori, M.; Tsukita, S.; Yamada, T.; et al. Metformin directly binds the alarmin HMGB1 and inhibits its proinflammatory activity. J. Biol. Chem. 2017, 292, 8436–8446. [Google Scholar] [CrossRef] [Green Version]
  31. Han, Y.; Yuan, F.; Deng, C.; He, F.; Zhang, Y.; Shen, H.; Chen, Z.; Qian, L. Metformin decreases LPS-induced inflammatory response in rabbit annulus fibrosus stem/progenitor cells by blocking HMGB1 release. Aging 2019, 11, 10252–10265. [Google Scholar] [CrossRef]
  32. Tsoyi, K.; Lee, T.Y.; Lee, Y.S.; Kim, H.J.; Seo, H.G.; Lee, J.H.; Chang, K.C. Heme-oxygenase-1 induction and carbon monoxide-releasing molecule inhibit lipopolysaccharide (LPS)-induced high-mobility group box 1 release in vitro and improve survival of mice in LPS- and cecal ligation and puncture-induced sepsis model in vivo. Mol. Pharmacol. 2009, 76, 173–182. [Google Scholar] [CrossRef] [Green Version]
  33. Wang, J.; Hu, X.; Jiang, H. Nrf-2-HO-1-HMGB1 axis: An important therapeutic approach for protection against myocardial ischemia and reperfusion injury. Int. J. Cardiol. 2014, 172, 223–224. [Google Scholar] [CrossRef] [PubMed]
  34. Rao, Z.; Zhang, N.; Xu, N.; Pan, Y.; Xiao, M.; Wu, J.; Zhou, H.; Yang, S.; Chen, Y. 1,25-Dihydroxyvitamin D Inhibits LPS-Induced High-Mobility Group Box 1 (HMGB1) Secretion via Targeting the NF-E2-Related Factor 2-Hemeoxygenase-1-HMGB1 Pathway in Macrophages. Front. Immunol. 2017, 8, 1308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Yu, Y.; Tang, D.; Kang, R. Oxidative stress-mediated HMGB1 biology. Front. Physiol. 2015, 6, 93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Scaffidi, P.; Misteli, T.; Bianchi, M.E. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature 2002, 418, 191–195. [Google Scholar] [CrossRef]
  37. Yanai, H.; Ban, T.; Wang, Z.; Choi, M.K.; Kawamura, T.; Negishi, H.; Nakasato, M.; Lu, Y.; Hangai, S.; Koshiba, R.; et al. HMGB proteins function as universal sentinels for nucleic-acid-mediated innate immune responses. Nature 2009, 462, 99–103. [Google Scholar] [CrossRef]
  38. Yang, H.; Wang, H.; Ju, Z.; Ragab, A.A.; Lundbäck, P.; Long, W.; Valdes-Ferrer, S.I.; He, M.; Pribis, J.P.; Li, J.; et al. MD-2 is required for disulfide HMGB1-dependent TLR4 signaling. J. Exp. Med. 2015, 212, 5–14. [Google Scholar] [CrossRef] [Green Version]
  39. Xu, J.; Jiang, Y.; Wang, J.; Shi, X.; Liu, Q.; Liu, Z.; Li, Y.; Scott, M.J.; Xiao, G.; Li, S.; et al. Macrophage endocytosis of high-mobility group box 1 triggers pyroptosis. Cell Death Differ. 2014, 21, 1229–1239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Andersson, U.; Tracey, K.J. HMGB1 is a therapeutic target for sterile inflammation and infection. Annu Rev. Immunol. 2011, 29, 139–162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Lu, M.; Yu, S.; Xu, W.; Gao, B.; Xiong, S. HMGB1 Promotes Systemic Lupus Erythematosus by Enhancing Macrophage Inflammatory Response. J. Immunol. Res. 2015, 2015, 946748. [Google Scholar] [CrossRef] [Green Version]
  42. Germain, R.N.; Meier-Schellersheim, M.; Nita-Lazar, A.; Fraser, I.D. Systems biology in immunology: A computational modeling perspective. Annu. Rev. Immunol. 2011, 29, 527–585. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Lundbäck, P.; Lea, J.D.; Sowinska, A.; Ottosson, L.; Fürst, C.M.; Steen, J.; Aulin, C.; Clarke, J.I.; Kipar, A.; Klevenvall, L.; et al. A novel high mobility group box 1 neutralizing chimeric antibody attenuates drug-induced liver injury and postinjury inflammation in mice. Hepatology 2016, 64, 1699–1710. [Google Scholar] [CrossRef] [Green Version]
  44. Watanabe, H.; Watanabe, K.S.; Liu, K.; Hiramatsu, S.; Zeggar, S.; Katsuyama, E.; Tatebe, N.; Akahoshi, A.; Takenaka, F.; Hanada, T.; et al. Anti-high Mobility Group Box 1 Antibody Ameliorates Albuminuria in MRL/lpr Lupus-Prone Mice. Mol. Ther. Methods Clin. Dev. 2017, 6, 31–39. [Google Scholar] [CrossRef] [Green Version]
  45. Schmidt, A.M.; Yan, S.D.; Yan, S.F.; Stern, D.M. The multiligand receptor RAGE as a progression factor amplifying immune and inflammatory responses. J. Clin. Invest. 2001, 108, 949–955. [Google Scholar] [CrossRef]
  46. Sessa, L.; Gatti, E.; Zeni, F.; Antonelli, A.; Catucci, A.; Koch, M.; Pompilio, G.; Fritz, G.; Raucci, A.; Bianchi, M.E. The receptor for advanced glycation end-products (RAGE) is only present in mammals, and belongs to a family of cell adhesion molecules (CAMs). PLoS ONE 2014, 9, e86903. [Google Scholar] [CrossRef]
  47. Constien, R.; Forde, A.; Liliensiek, B.; Gröne, H.J.; Nawroth, P.; Hämmerling, G.; Arnold, B. Characterization of a novel EGFP reporter mouse to monitor Cre recombination as demonstrated by a Tie2 Cre mouse line. Genesis 2001, 30, 36–44. [Google Scholar] [CrossRef]
  48. Myint, K.M.; Yamamoto, Y.; Doi, T.; Kato, I.; Harashima, A.; Yonekura, H.; Watanabe, T.; Shinohara, H.; Takeuchi, M.; Tsuneyama, K.; et al. RAGE control of diabetic nephropathy in a mouse model: Effects of RAGE gene disruption and administration of low-molecular weight heparin. Diabetes 2006, 55, 2510–2522. [Google Scholar] [CrossRef] [Green Version]
  49. Galbiati, F.; Razani, B.; Lisanti, M.P. Emerging themes in lipid rafts and caveolae. Cell 2001, 106, 403–411. [Google Scholar] [CrossRef] [Green Version]
  50. Sirois, C.M.; Jin, T.; Miller, A.L.; Bertheloot, D.; Nakamura, H.; Horvath, G.L.; Mian, A.; Jiang, J.; Schrum, J.; Bossaller, L.; et al. RAGE is a nucleic acid receptor that promotes inflammatory responses to DNA. J. Exp. Med. 2013, 210, 2447–2463. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Hudson, B.I.; Lippman, M.E. Targeting RAGE Signaling in Inflammatory Disease. Annu. Rev. Med. 2018, 69, 349–364. [Google Scholar] [CrossRef] [PubMed]
  52. Sakaguchi, M.; Kinoshita, R.; Putranto, E.W.; Ruma, I.M.W.; Sumardika, I.W.; Youyi, C.; Tomonobu, N.; Yamamoto, K.I.; Murata, H. Signal Diversity of Receptor for Advanced Glycation End Products. Acta Med. Okayama 2017, 71, 459–465. [Google Scholar] [CrossRef] [PubMed]
  53. Jules, J.; Maiguel, D.; Hudson, B.I. Alternative splicing of the RAGE cytoplasmic domain regulates cell signaling and function. PLoS ONE 2013, 8, e78267. [Google Scholar] [CrossRef] [Green Version]
  54. Xie, J.; Reverdatto, S.; Frolov, A.; Hoffmann, R.; Burz, D.S.; Shekhtman, A. Structural basis for pattern recognition by the receptor for advanced glycation end products (RAGE). J. Biol. Chem. 2008, 283, 27255–27269. [Google Scholar] [CrossRef] [Green Version]
  55. Kierdorf, K.; Fritz, G. RAGE regulation and signaling in inflammation and beyond. J. Leukoc. Biol. 2013, 94, 55–68. [Google Scholar] [CrossRef] [PubMed]
  56. Sakaguchi, M.; Murata, H.; Aoyama, Y.; Hibino, T.; Putranto, E.W.; Ruma, I.M.; Inoue, Y.; Sakaguchi, Y.; Yamamoto, K.; Kinoshita, R.; et al. DNAX-activating protein 10 (DAP10) membrane adaptor associates with receptor for advanced glycation end products (RAGE) and modulates the RAGE-triggered signaling pathway in human keratinocytes. J. Biol. Chem. 2014, 289, 23389–23402. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Hudson, B.I.; Carter, A.M.; Harja, E.; Kalea, A.Z.; Arriero, M.; Yang, H.; Grant, P.J.; Schmidt, A.M. Identification, classification, and expression of RAGE gene splice variants. FASEB J. 2008, 22, 1572–1580. [Google Scholar] [CrossRef]
  58. Raucci, A.; Cugusi, S.; Antonelli, A.; Barabino, S.M.; Monti, L.; Bierhaus, A.; Reiss, K.; Saftig, P.; Bianchi, M.E. A soluble form of the receptor for advanced glycation endproducts (RAGE) is produced by proteolytic cleavage of the membrane-bound form by the sheddase a disintegrin and metalloprotease 10 (ADAM10). FASEB J. 2008, 22, 3716–3727. [Google Scholar] [CrossRef] [PubMed]
  59. Hanford, L.E.; Enghild, J.J.; Valnickova, Z.; Petersen, S.V.; Schaefer, L.M.; Schaefer, T.M.; Reinhart, T.A.; Oury, T.D. Purification and characterization of mouse soluble receptor for advanced glycation end products (sRAGE). J. Biol. Chem. 2004, 279, 50019–50024. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Hori, O.; Brett, J.; Slattery, T.; Cao, R.; Zhang, J.; Chen, J.X.; Nagashima, M.; Lundh, E.R.; Vijay, S.; Nitecki, D. The receptor for advanced glycation end products (RAGE) is a cellular binding site for amphoterin. Mediation of neurite outgrowth and co-expression of rage and amphoterin in the developing nervous system. J. Biol. Chem. 1995, 270, 25752–25761. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Huttunen, H.J.; Fages, C.; Kuja-Panula, J.; Ridley, A.J.; Rauvala, H. Receptor for advanced glycation end products-binding COOH-terminal motif of amphoterin inhibits invasive migration and metastasis. Cancer Res. 2002, 62, 4805–4811. [Google Scholar] [PubMed]
  62. Taguchi, A.; Blood, D.C.; del Toro, G.; Canet, A.; Lee, D.C.; Qu, W.; Tanji, N.; Lu, Y.; Lalla, E.; Fu, C.; et al. Blockade of RAGE-amphoterin signalling suppresses tumour growth and metastases. Nature 2000, 405, 354–360. [Google Scholar] [CrossRef]
  63. Sakaguchi, M.; Murata, H.; Yamamoto, K.; Ono, T.; Sakaguchi, Y.; Motoyama, A.; Hibino, T.; Kataoka, K.; Huh, N.H. TIRAP, an adaptor protein for TLR2/4, transduces a signal from RAGE phosphorylated upon ligand binding. PLoS ONE 2011, 6, e23132. [Google Scholar] [CrossRef] [Green Version]
  64. Kang, R.; Tang, D.; Schapiro, N.E.; Loux, T.; Livesey, K.M.; Billiar, T.R.; Wang, H.; Van Houten, B.; Lotze, M.T.; Zeh, H.J. The HMGB1/RAGE inflammatory pathway promotes pancreatic tumor growth by regulating mitochondrial bioenergetics. Oncogene 2014, 33, 567–577. [Google Scholar] [CrossRef] [Green Version]
  65. Huebener, P.; Pradere, J.P.; Hernandez, C.; Gwak, G.Y.; Caviglia, J.M.; Mu, X.; Loike, J.D.; Jenkins, R.E.; Antoine, D.J.; Schwabe, R.F. The HMGB1/RAGE axis triggers neutrophil-mediated injury amplification following necrosis. J. Clin. Invest. 2015, 125, 539–550. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Flyvbjerg, A.; Denner, L.; Schrijvers, B.F.; Tilton, R.G.; Mogensen, T.H.; Paludan, S.R.; Rasch, R. Long-term renal effects of a neutralizing RAGE antibody in obese type 2 diabetic mice. Diabetes 2004, 53, 166–172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Johnson, L.L.; Johnson, J.; Ober, R.; Holland, A.; Zhang, G.; Backer, M.; Backer, J.; Ali, Z.; Tekabe, Y. Novel Receptor for Advanced Glycation End Products-Blocking Antibody to Treat Diabetic Peripheral Artery Disease. J. Am. Heart Assoc. 2021, 10, e016696. [Google Scholar] [CrossRef] [PubMed]
  68. Christaki, E.; Opal, S.M.; Keith, J.C.; Kessimian, N.; Palardy, J.E.; Parejo, N.A.; Tan, X.Y.; Piche-Nicholas, N.; Tchistiakova, L.; Vlasuk, G.P.; et al. A monoclonal antibody against RAGE alters gene expression and is protective in experimental models of sepsis and pneumococcal pneumonia. Shock 2011, 35, 492–498. [Google Scholar] [CrossRef] [PubMed]
  69. Lee, S.W.; Park, K.H.; Park, S.; Kim, J.H.; Hong, S.Y.; Lee, S.K.; Choi, D.; Park, Y.B. Soluble receptor for advanced glycation end products alleviates nephritis in (NZB/NZW)F1 mice. Arthritis Rheum. 2013, 65, 1902–1912. [Google Scholar] [CrossRef] [PubMed]
  70. Burstein, A.H.; Grimes, I.; Galasko, D.R.; Aisen, P.S.; Sabbagh, M.; Mjalli, A.M. Effect of TTP488 in patients with mild to moderate Alzheimer’s disease. BMC Neurol. 2014, 14, 12. [Google Scholar] [CrossRef] [Green Version]
  71. Galasko, D.; Bell, J.; Mancuso, J.Y.; Kupiec, J.W.; Sabbagh, M.N.; van Dyck, C.; Thomas, R.G.; Aisen, P.S.; Study, A.s.D.C. Clinical trial of an inhibitor of RAGE-Aβ interactions in Alzheimer disease. Neurology 2014, 82, 1536–1542. [Google Scholar] [CrossRef]
  72. Deane, R.; Singh, I.; Sagare, A.P.; Bell, R.D.; Ross, N.T.; LaRue, B.; Love, R.; Perry, S.; Paquette, N.; Deane, R.J.; et al. A multimodal RAGE-specific inhibitor reduces amyloid β-mediated brain disorder in a mouse model of Alzheimer disease. J. Clin. Invest. 2012, 122, 1377–1392. [Google Scholar] [CrossRef] [Green Version]
  73. Yang, F.; Wang, Z.; Zhang, J.H.; Tang, J.; Liu, X.; Tan, L.; Huang, Q.Y.; Feng, H. Receptor for advanced glycation end-product antagonist reduces blood-brain barrier damage after intracerebral hemorrhage. Stroke 2015, 46, 1328–1336. [Google Scholar] [CrossRef]
  74. Lee, H.; Park, J.R.; Kim, W.J.; Sundar, I.K.; Rahman, I.; Park, S.M.; Yang, S.R. Blockade of RAGE ameliorates elastase-induced emphysema development and progression. FASEB J. 2017, 31, 2076–2089. [Google Scholar] [CrossRef] [Green Version]
  75. Kwak, T.; Drews-Elger, K.; Ergonul, A.; Miller, P.C.; Braley, A.; Hwang, G.H.; Zhao, D.; Besser, A.; Yamamoto, Y.; Yamamoto, H.; et al. Targeting of RAGE-ligand signaling impairs breast cancer cell invasion and metastasis. Oncogene 2017, 36, 1559–1572. [Google Scholar] [CrossRef]
  76. Body-Malapel, M.; Djouina, M.; Waxin, C.; Langlois, A.; Gower-Rousseau, C.; Zerbib, P.; Schmidt, A.M.; Desreumaux, P.; Boulanger, E.; Vignal, C. The RAGE signaling pathway is involved in intestinal inflammation and represents a promising therapeutic target for Inflammatory Bowel Diseases. Mucosal Immunol. 2019, 12, 468–478. [Google Scholar] [CrossRef] [PubMed]
  77. Shen, L.; Zhang, T.; Yang, Y.; Lu, D.; Xu, A.; Li, K. FPS-ZM1 Alleviates Neuroinflammation in Focal Cerebral Ischemia Rats via Blocking Ligand/RAGE/DIAPH1 Pathway. ACS Chem. Neurosci. 2021, 12, 63–78. [Google Scholar] [CrossRef] [PubMed]
  78. Matsui, T.; Higashimoto, Y.; Nishino, Y.; Nakamura, N.; Fukami, K.; Yamagishi, S.I. RAGE-Aptamer Blocks the Development and Progression of Experimental Diabetic Nephropathy. Diabetes 2017, 66, 1683–1695. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Nakamura, N.; Matsui, T.; Ishibashi, Y.; Sotokawauchi, A.; Fukami, K.; Higashimoto, Y.; Yamagishi, S.I. RAGE-aptamer Attenuates the Growth and Liver Metastasis of Malignant Melanoma in Nude Mice. Mol. Med. 2017, 23, 295–306. [Google Scholar] [CrossRef]
  80. Tian, J.; Avalos, A.M.; Mao, S.Y.; Chen, B.; Senthil, K.; Wu, H.; Parroche, P.; Drabic, S.; Golenbock, D.; Sirois, C.; et al. Toll-like receptor 9-dependent activation by DNA-containing immune complexes is mediated by HMGB1 and RAGE. Nat. Immunol. 2007, 8, 487–496. [Google Scholar] [CrossRef]
  81. Porat, A.; Giat, E.; Kowal, C.; He, M.; Son, M.; Latz, E.; Ben-Zvi, I.; Al-Abed, Y.; Diamond, B. DNA-Mediated Interferon Signature Induction by SLE Serum Occurs in Monocytes Through Two Pathways: A Mechanism to Inhibit Both Pathways. Front. Immunol. 2018, 9, 2824. [Google Scholar] [CrossRef]
  82. Qing, X.; Pitashny, M.; Thomas, D.B.; Barrat, F.J.; Hogarth, M.P.; Putterman, C. Pathogenic anti-DNA antibodies modulate gene expression in mesangial cells: Involvement of HMGB1 in anti-DNA antibody-induced renal injury. Immunol. Lett. 2008, 121, 61–73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Ma, C.Y.; Ma, J.L.; Jiao, Y.L.; Li, J.F.; Wang, L.C.; Yang, Q.R.; You, L.; Cui, B.; Chen, Z.J.; Zhao, Y.R. The plasma level of soluble receptor for advanced glycation end products is decreased in patients with systemic lupus erythematosus. Scand. J. Immunol. 2012, 75, 614–622. [Google Scholar] [CrossRef]
  84. Lan, L.; Han, F.; Lang, X.; Chen, J. Monocyte Chemotactic Protein-1, Fractalkine, and Receptor for Advanced Glycation End Products in Different Pathological Types of Lupus Nephritis and Their Value in Different Treatment Prognoses. PLoS ONE 2016, 11, e0159964. [Google Scholar] [CrossRef]
  85. Goury, A.; Meghraoui-Kheddar, A.; Belmokhtar, K.; Vuiblet, V.; Ortillon, J.; Jaisson, S.; Devy, J.; Le Naour, R.; Tabary, T.; Cohen, J.H.; et al. Deletion of receptor for advanced glycation end products exacerbates lymphoproliferative syndrome and lupus nephritis in B6-MRL Fas lpr/j mice. J. Immunol. 2015, 194, 3612–3622. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Wibisono, D.; Csernok, E.; Lamprecht, P.; Holle, J.U.; Gross, W.L.; Moosig, F. Serum HMGB1 levels are increased in active Wegener’s granulomatosis and differentiate between active forms of ANCA-associated vasculitis. Ann. Rheum. Dis. 2010, 69, 1888–1889. [Google Scholar] [CrossRef] [PubMed]
  87. Bruchfeld, A.; Wendt, M.; Bratt, J.; Qureshi, A.R.; Chavan, S.; Tracey, K.J.; Palmblad, K.; Gunnarsson, I. High-mobility group box-1 protein (HMGB1) is increased in antineutrophilic cytoplasmatic antibody (ANCA)-associated vasculitis with renal manifestations. Mol. Med. 2011, 17, 29–35. [Google Scholar] [CrossRef] [PubMed]
  88. Wang, C.; Wang, H.; Chang, D.Y.; Hao, J.; Zhao, M.H.; Chen, M. High mobility group box 1 contributes to anti-neutrophil cytoplasmic antibody-induced neutrophils activation through receptor for advanced glycation end products (RAGE) and Toll-like receptor 4. Arthritis Res. 2015, 17, 64. [Google Scholar] [CrossRef] [Green Version]
  89. Nakazawa, D.; Tomaru, U.; Suzuki, A.; Masuda, S.; Hasegawa, R.; Kobayashi, T.; Nishio, S.; Kasahara, M.; Ishizu, A. Abnormal conformation and impaired degradation of propylthiouracil-induced neutrophil extracellular traps: Implications of disordered neutrophil extracellular traps in a rat model of myeloperoxidase antineutrophil cytoplasmic antibody-associated vasculitis. Arthritis Rheum. 2012, 64, 3779–3787. [Google Scholar] [CrossRef]
  90. Nakazawa, D.; Tomaru, U.; Yamamoto, C.; Jodo, S.; Ishizu, A. Abundant neutrophil extracellular traps in thrombus of patient with microscopic polyangiitis. Front. Immunol. 2012, 3, 333. [Google Scholar] [CrossRef] [Green Version]
  91. Kessenbrock, K.; Krumbholz, M.; Schönermarck, U.; Back, W.; Gross, W.L.; Werb, Z.; Gröne, H.J.; Brinkmann, V.; Jenne, D.E. Netting neutrophils in autoimmune small-vessel vasculitis. Nat. Med. 2009, 15, 623–625. [Google Scholar] [CrossRef]
  92. Fuchs, T.A.; Brill, A.; Duerschmied, D.; Schatzberg, D.; Monestier, M.; Myers, D.D., Jr.; Wrobleski, S.K.; Wakefield, T.W.; Hartwig, J.H.; Wagner, D.D. Extracellular DNA traps promote thrombosis. Proc. Natl. Acad. Sci. USA 2010, 107, 15880–15885. [Google Scholar] [CrossRef] [Green Version]
  93. Ma, Y.H.; Ma, T.T.; Wang, C.; Wang, H.; Chang, D.Y.; Chen, M.; Zhao, M.H. High-mobility group box 1 potentiates antineutrophil cytoplasmic antibody-inducing neutrophil extracellular traps formation. Arthritis Res. 2016, 18, 2. [Google Scholar] [CrossRef] [Green Version]
  94. Maugeri, N.; Campana, L.; Gavina, M.; Covino, C.; De Metrio, M.; Panciroli, C.; Maiuri, L.; Maseri, A.; D’Angelo, A.; Bianchi, M.E.; et al. Activated platelets present high mobility group box 1 to neutrophils, inducing autophagy and promoting the extrusion of neutrophil extracellular traps. J. Thromb. Haemost. JTH 2014, 12, 2074–2088. [Google Scholar] [CrossRef]
  95. Peschel, A.; Basu, N.; Benharkou, A.; Brandes, R.; Brown, M.; Rees, A.J.; Kain, R. Autoantibodies to hLAMP-2 in ANCA-negative pauci-immune focal necrotizing GN. J. Am. Soc. Nephrol. 2014, 25, 455–463. [Google Scholar] [CrossRef] [Green Version]
  96. Saftig, P.; Klumperman, J. Lysosome biogenesis and lysosomal membrane proteins: Trafficking meets function. Nat. Rev. Mol. Cell Biol. 2009, 10, 623–635. [Google Scholar] [CrossRef] [PubMed]
  97. Leone, D.A.; Peschel, A.; Brown, M.; Schachner, H.; Ball, M.J.; Gyuraszova, M.; Salzer-Muhar, U.; Fukuda, M.; Vizzardelli, C.; Bohle, B.; et al. Surface LAMP-2 Is an Endocytic Receptor That Diverts Antigen Internalized by Human Dendritic Cells into Highly Immunogenic Exosomes. J. Immunol. 2017, 199, 531–546. [Google Scholar] [CrossRef]
  98. Boone, B.A.; Orlichenko, L.; Schapiro, N.E.; Loughran, P.; Gianfrate, G.C.; Ellis, J.T.; Singhi, A.D.; Kang, R.; Tang, D.; Lotze, M.T.; et al. The receptor for advanced glycation end products (RAGE) enhances autophagy and neutrophil extracellular traps in pancreatic cancer. Cancer Gene 2015, 22, 326–334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Zhu, X.; Messer, J.S.; Wang, Y.; Lin, F.; Cham, C.M.; Chang, J.; Billiar, T.R.; Lotze, M.T.; Boone, D.L.; Chang, E.B. Cytosolic HMGB1 controls the cellular autophagy/apoptosis checkpoint during inflammation. J. Clin. Invest. 2015, 125, 1098–1110. [Google Scholar] [CrossRef] [Green Version]
  100. Wang, J.; Fu, L.; Yang, H.; Cao, K.; Sun, Q.; Chen, T. The Anti-inflammatory Effects of HMGB1 Blockades in a Mouse Model of Cutaneous Vasculitis. Front. Immunol. 2020, 11, 2032. [Google Scholar] [CrossRef] [PubMed]
  101. Watanabe, T.; Kubota, S.; Nagaya, M.; Ozaki, S.; Nagafuchi, H.; Akashi, K.; Taira, Y.; Tsukikawa, S.; Oowada, S.; Nakano, S. The role of HMGB-1 on the development of necrosis during hepatic ischemia and hepatic ischemia/reperfusion injury in mice. J. Surg. Res. 2005, 124, 59–66. [Google Scholar] [CrossRef]
  102. Tsung, A.; Sahai, R.; Tanaka, H.; Nakao, A.; Fink, M.P.; Lotze, M.T.; Yang, H.; Li, J.; Tracey, K.J.; Geller, D.A.; et al. The nuclear factor HMGB1 mediates hepatic injury after murine liver ischemia-reperfusion. J. Exp. Med. 2005, 201, 1135–1143. [Google Scholar] [CrossRef] [PubMed]
  103. Kim, J.B.; Sig Choi, J.; Yu, Y.M.; Nam, K.; Piao, C.S.; Kim, S.W.; Lee, M.H.; Han, P.L.; Park, J.S.; Lee, J.K. HMGB1, a novel cytokine-like mediator linking acute neuronal death and delayed neuroinflammation in the postischemic brain. J. Neurosci. Off. J. Soc. Neurosci. 2006, 26, 6413–6421. [Google Scholar] [CrossRef] [Green Version]
  104. Hayakawa, K.; Pham, L.D.; Katusic, Z.S.; Arai, K.; Lo, E.H. Astrocytic high-mobility group box 1 promotes endothelial progenitor cell-mediated neurovascular remodeling during stroke recovery. Proc. Natl. Acad. Sci. USA 2012, 109, 7505–7510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Khan, M.I.; Rath, S.; Adhami, V.M.; Mukhtar, H. Hypoxia driven glycation: Mechanisms and therapeutic opportunities. Semin. Cancer Biol. 2018, 49, 75–82. [Google Scholar] [CrossRef] [PubMed]
  106. Huber, R.; Meier, B.; Otsuka, A.; Fenini, G.; Satoh, T.; Gehrke, S.; Widmer, D.; Levesque, M.P.; Mangana, J.; Kerl, K.; et al. Tumour hypoxia promotes melanoma growth and metastasis via High Mobility Group Box-1 and M2-like macrophages. Sci. Rep. 2016, 6, 29914. [Google Scholar] [CrossRef] [Green Version]
  107. Pichiule, P.; Chavez, J.C.; Schmidt, A.M.; Vannucci, S.J. Hypoxia-inducible factor-1 mediates neuronal expression of the receptor for advanced glycation end products following hypoxia/ischemia. J. Biol. Chem. 2007, 282, 36330–36340. [Google Scholar] [CrossRef] [Green Version]
  108. Chang, J.S.; Wendt, T.; Qu, W.; Kong, L.; Zou, Y.S.; Schmidt, A.M.; Yan, S.F. Oxygen deprivation triggers upregulation of early growth response-1 by the receptor for advanced glycation end products. Circ. Res. 2008, 102, 905–913. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Wang, J.; Yang, H.; Hu, X.; Fu, W.; Xie, J.; Zhou, X.; Xu, W.; Jiang, H. Dobutamine-mediated heme oxygenase-1 induction via PI3K and p38 MAPK inhibits high mobility group box 1 protein release and attenuates rat myocardial ischemia/reperfusion injury in vivo. J. Surg. Res. 2013, 183, 509–516. [Google Scholar] [CrossRef]
  110. Seo, M.S.; Kim, H.J.; Kim, H.; Park, S.W. Ethyl Pyruvate Directly Attenuates Active Secretion of HMGB1 in Proximal Tubular Cells via Induction of Heme Oxygenase-1. J. Clin. Med. 2019, 8, 629. [Google Scholar] [CrossRef] [Green Version]
  111. Slowik, A.; Merres, J.; Elfgen, A.; Jansen, S.; Mohr, F.; Wruck, C.J.; Pufe, T.; Brandenburg, L.O. Involvement of formyl peptide receptors in receptor for advanced glycation end products (RAGE)--and amyloid beta 1-42-induced signal transduction in glial cells. Mol. Neurodegener. 2012, 7, 55. [Google Scholar] [CrossRef] [Green Version]
  112. Ichiki, T.; Koga, T.; Okuno, T.; Saeki, K.; Yamamoto, Y.; Yamamoto, H.; Sakaguchi, M.; Yokomizo, T. Modulation of leukotriene B4 receptor 1 signaling by receptor for advanced glycation end products (RAGE). FASEB J. 2016, 30, 1811–1822. [Google Scholar] [CrossRef] [Green Version]
  113. Rojas, A.; Delgado-López, F.; Perez-Castro, R.; Gonzalez, I.; Romero, J.; Rojas, I.; Araya, P.; Añazco, C.; Morales, E.; Llanos, J. HMGB1 enhances the protumoral activities of M2 macrophages by a RAGE-dependent mechanism. Tumour Biol. J. Int. Soc. Oncodevelop. Biol. Med. 2016, 37, 3321–3329. [Google Scholar] [CrossRef] [PubMed]
  114. Liao, Y.; Liu, S.; Fu, S.; Wu, J. HMGB1 in Radiotherapy: A Two Headed Signal Regulating Tumor Radiosensitivity and Immunity. Oncotargets Ther. 2020, 13, 6859–6871. [Google Scholar] [CrossRef] [PubMed]
  115. Son, M.; Porat, A.; He, M.; Suurmond, J.; Santiago-Schwarz, F.; Andersson, U.; Coleman, T.R.; Volpe, B.T.; Tracey, K.J.; Al-Abed, Y.; et al. C1q and HMGB1 reciprocally regulate human macrophage polarization. Blood 2016, 128, 2218–2228. [Google Scholar] [CrossRef] [Green Version]
  116. Das, U.N. Arachidonic acid and lipoxin A4 as possible endogenous anti-diabetic molecules. Prostaglandins Leukot. Essent. Fat. Acids 2013, 88, 201–210. [Google Scholar] [CrossRef]
  117. Levy, B.D.; Clish, C.B.; Schmidt, B.; Gronert, K.; Serhan, C.N. Lipid mediator class switching during acute inflammation: Signals in resolution. Nat. Immunol. 2001, 2, 612–619. [Google Scholar] [CrossRef]
  118. Liu, T.; Xiang, A.; Peng, T.; Doran, A.C.; Tracey, K.J.; Barnes, B.J.; Tabas, I.; Son, M.; Diamond, B. HMGB1-C1q complexes regulate macrophage function by switching between leukotriene and specialized proresolving mediator biosynthesis. Proc. Natl. Acad. Sci. USA 2019, 116, 23254–23263. [Google Scholar] [CrossRef]
  119. Wild, C.A.; Bergmann, C.; Fritz, G.; Schuler, P.; Hoffmann, T.K.; Lotfi, R.; Westendorf, A.; Brandau, S.; Lang, S. HMGB1 conveys immunosuppressive characteristics on regulatory and conventional T cells. Int. Immunol. 2012, 24, 485–494. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Yang, H.; Wang, H.; Levine, Y.A.; Gunasekaran, M.K.; Wang, Y.; Addorisio, M.; Zhu, S.; Li, W.; Li, J.; de Kleijn, D.P.; et al. Identification of CD163 as an antiinflammatory receptor for HMGB1-haptoglobin complexes. JCI Insight 2016, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Bandala-Sanchez, E.; Bediaga, N.G.; Goddard-Borger, E.D.; Ngui, K.; Naselli, G.; Stone, N.L.; Neale, A.M.; Pearce, L.A.; Wardak, A.; Czabotar, P.; et al. CD52 glycan binds the proinflammatory B box of HMGB1 to engage the Siglec-10 receptor and suppress human T cell function. Proc. Natl. Acad. Sci. USA 2018, 115, 7783–7788. [Google Scholar] [CrossRef] [Green Version]
  122. Gabrilovich, D.I.; Bronte, V.; Chen, S.H.; Colombo, M.P.; Ochoa, A.; Ostrand-Rosenberg, S.; Schreiber, H. The terminology issue for myeloid-derived suppressor cells. Cancer Res. 2007, 67, 425. [Google Scholar] [CrossRef] [Green Version]
  123. Parker, K.H.; Sinha, P.; Horn, L.A.; Clements, V.K.; Yang, H.; Li, J.; Tracey, K.J.; Ostrand-Rosenberg, S. HMGB1 enhances immune suppression by facilitating the differentiation and suppressive activity of myeloid-derived suppressor cells. Cancer Res. 2014, 74, 5723–5733. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. The regulation of HMGB1 secretion. Passive release of HMGB1 involves the plasma membrane disruption through cell death mechanisms. Inflammation and immune activation induce HMGB1 secretion. Post-translational modifications-mediated active secretion of HMGB1 occurs via secretory lysosomes. C5a and C5aR2 pathway induces HMGB1 release. Small molecules such as glycyrrhizin and metformin inhibit nuclear HMGB1 translocation to the cytosol. The nuclear factor-erythroid 2-related factor 2 (Nrf2)/hemeoxygenase-1 (HO-1) pathway also suppresses the translocation and secretion of HMGB1.
Figure 1. The regulation of HMGB1 secretion. Passive release of HMGB1 involves the plasma membrane disruption through cell death mechanisms. Inflammation and immune activation induce HMGB1 secretion. Post-translational modifications-mediated active secretion of HMGB1 occurs via secretory lysosomes. C5a and C5aR2 pathway induces HMGB1 release. Small molecules such as glycyrrhizin and metformin inhibit nuclear HMGB1 translocation to the cytosol. The nuclear factor-erythroid 2-related factor 2 (Nrf2)/hemeoxygenase-1 (HO-1) pathway also suppresses the translocation and secretion of HMGB1.
Cells 10 00564 g001
Figure 2. RAGE structure and signaling pathway. The receptor for advanced glycation endproducts (RAGE) has three extracellular immunoglobulin-like domains (V, C1, and C2), a single transmembrane helix, and a C-terminal short domain, and exists in lipid rafts. Soluble RAGE (sRAGE) is created by alternative splicing or cleaved by protease and functions as a decoy receptor. When binding with high mobility group box 1 (HMGB1) and nucleic acid, RAGE internalizes into the cytosol and interacts with Toll-like receptors (TLRs). RAGE transduces down-stream signaling upon binding with ligands and adaptor proteins, including diaphanous homolog 1 (Diaph1), toll-interleukin 1 receptor domain-containing adaptor protein (TIRAP), and myeloid differentiation primary response 88 (MyD88), resulting in type 1 interferon and pro-inflammatory cytokines production.
Figure 2. RAGE structure and signaling pathway. The receptor for advanced glycation endproducts (RAGE) has three extracellular immunoglobulin-like domains (V, C1, and C2), a single transmembrane helix, and a C-terminal short domain, and exists in lipid rafts. Soluble RAGE (sRAGE) is created by alternative splicing or cleaved by protease and functions as a decoy receptor. When binding with high mobility group box 1 (HMGB1) and nucleic acid, RAGE internalizes into the cytosol and interacts with Toll-like receptors (TLRs). RAGE transduces down-stream signaling upon binding with ligands and adaptor proteins, including diaphanous homolog 1 (Diaph1), toll-interleukin 1 receptor domain-containing adaptor protein (TIRAP), and myeloid differentiation primary response 88 (MyD88), resulting in type 1 interferon and pro-inflammatory cytokines production.
Cells 10 00564 g002
Figure 3. RAGE function as heterodimers. The functional interaction between RAGE and DNAX-activating protein 10 (DAP10) coordinately regulates S100-mediated cell survival. Formyl-peptide-receptors (FPRs) reacts with the broad ligand spectrum through the interaction with RAGE. Leukotriene B4 (LTB4) receptor 1 (BLT1) interacts with RAGE and induces proinflammatory cytokines and chemokines. HMGB1 promotes leukotriene production, induces interferon regulatory factor 5 in a RAGE-dependent manner. C1q can form a multimolecular signaling complex with HMGB1, RAGE, and Leukocyte-Associated Ig-like Receptor-1 (LAIR-1) and produces specialized pro-resolving lipid mediators (SPMs) and promotes M2-like macrophage polarization, which contributes to the resolution of inflammation.
Figure 3. RAGE function as heterodimers. The functional interaction between RAGE and DNAX-activating protein 10 (DAP10) coordinately regulates S100-mediated cell survival. Formyl-peptide-receptors (FPRs) reacts with the broad ligand spectrum through the interaction with RAGE. Leukotriene B4 (LTB4) receptor 1 (BLT1) interacts with RAGE and induces proinflammatory cytokines and chemokines. HMGB1 promotes leukotriene production, induces interferon regulatory factor 5 in a RAGE-dependent manner. C1q can form a multimolecular signaling complex with HMGB1, RAGE, and Leukocyte-Associated Ig-like Receptor-1 (LAIR-1) and produces specialized pro-resolving lipid mediators (SPMs) and promotes M2-like macrophage polarization, which contributes to the resolution of inflammation.
Cells 10 00564 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Watanabe, H.; Son, M. The Immune Tolerance Role of the HMGB1-RAGE Axis. Cells 2021, 10, 564. https://doi.org/10.3390/cells10030564

AMA Style

Watanabe H, Son M. The Immune Tolerance Role of the HMGB1-RAGE Axis. Cells. 2021; 10(3):564. https://doi.org/10.3390/cells10030564

Chicago/Turabian Style

Watanabe, Haruki, and Myoungsun Son. 2021. "The Immune Tolerance Role of the HMGB1-RAGE Axis" Cells 10, no. 3: 564. https://doi.org/10.3390/cells10030564

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop