AJA Asian Journal of Anesthesiology

Advancing, Capability, Improving lives

Review Article
Volume 51, Issue 1, Pages 28-33
Paladd Asavarut 1 , Hailin Zhao 1 , Jianteng Gu 2 , Daqing Ma 1
2682 Views


Abstract

HMGB1 is a chromosome-binding protein that also acts as a damage-associated molecular pattern molecule. It has potent proinflammatory effects and is one of key mediators of organ injury. Evidence from research has revealed its involvement in the signaling mechanisms of Toll-like receptors and the receptor for advanced glycation end-products in organ injury. HMGB1-mediated organ injuries are acute damage including ischemic, mechanical, allograft rejection and toxicity, and chronic diseases of the heart, kidneys, lungs, and brain. Strategies against HMGB1 and its associated cellular signal pathways need to be developed and may have preventive and therapeutic potentials in organ injury.

Keywords

HMGB1 prote; ininflammation; organ injuries;


1. Introduction

The high-mobility group box 1 protein (HMGB1) is a ubiquitously expressed nuclear factor and an important mediator of inflammation via receptors of the innate immune system. The inflammatory nature of organ and tissue injury from chemical or physical stress makes HMGB1 a crucial factor in the pathogenesis of acute and chronic diseases and injury. HMGB1, also a universal nucleic acid sensor, is able to detect exogenous nucleic acids in both the extracellular and intracellular spaces,1, 2 activating Toll-like receptors (TLRs). Its interaction with other small molecules such as p53 in a Beclin-1-dependent manner to modulate cell longevity has also been a subject of interest in cancer research,3 although this is beyond the scope of this review. Specifically, we focus here on and summarize the role of HMGB1 in acute and chronic organ injuries.

2. HMGB1 and inflammation

HMGB1 signals for the production of proinflammatory cytokines and chemokines though TLRs and the receptor for advanced glycation end-products (RAGE). Being present both intracellularly and extracellularly, HMGB1 triggers inflammation through direct binding to TLR4, or by formation of complexes with exogenous or endogenous molecules in the cytoplasm or extracellular space.45 The location and post-translational modification state of HMGB1 determine its physiologic action. Under normal conditions, it is responsible for maintenance of the chromosomal architecture, regulating processes in the genome.6 HMGB1 is trafficked into vesicles via a nonclassical pathway, awaiting caspase-1-dependent secretion when immunologic cells become activated.7 In other cells, it is an alarmin, a signal for damaged self.

In the event of infectious or sterile damage, the immune response is triggered primarily by pathogen-associated molecular pattern (PAMP) or damage-associated molecular pattern (DAMP) molecules. These rely on transduction by PAMP/DAMP receptors, resulting in the expression of proinflammatory cytokines such as tumor necrosis factor-alpha (TNF-α), interferon-gamma (IFN-γ), and interleukin (IL) IL-1β and IL-6.8 The nature of HMGB1-dependent TNF induction and release is different, as it is monophasic in response to endotoxic insult, but biphasic if mediated by HMGB1.9

The primary target of extracellular HMGB1 is TLR4, leading to the translocation of nuclear factor kappa B (NFκB) to the nucleus, and activation of interferon regulatory factor 3 (IRF3) and activator protein 1 (AP-1) to produce the inflammatory cytokine repertoire. It is said that HMGB1 is a late mediator of infectious damage, but an early one of sterile damage. Upon immune cell activation, HMGB1 expression and secretion takes 8–12 hours.10 The positive feedforward nature of immunologic signaling by HMGB1 is intended to prime and amplify the immune response through cytokine induction during stress, infection, or hypoxia.11 The induction of chemokines by immunocytes during this inflammatory process is stromal cell-derived factor-1 (CXCL12) dependent.12 In addition to other TLRs (2 and 9) that can also be activated by HMGB1, RAGE is another of its targets that is able to mediate chemotaxis, cell growth, differentiation, and its own upregulation.13 Despite its strong proinflammatory effect, its role in suppressing inflammation by interacting with sialic acid-binding immunoglobulin-like lectin 12 (Siglec12) in CD24+ immunocytes to prevent NFκB translocation to the nucleus has also been studied.14

3. Inflammation and organ injury

Damage can originate from internal and external events, resulting in mechanical or chemical/molecular stress, and triggering an inflammatory response. Despite initiating recovery and healing, inflammation causes organ injury by processes such as tissue destruction, remodeling, fibrosis, and others. Current literature indicates that HMGB1 plays an important role in mediating organ injury. For example, the marked increase in HMGB1 expression, both locally and/or systemically depending on the disease, has been documented. Damage caused applies to both acute and chronic diseases, and is proportional the amount of HMGB1 secreted.10

4. Current understanding of HMGB1-mediated vital organ injury

4.1. Ischemia and reperfusion injury

Ischemia/reperfusion (I/R) injury is characterized by an inflammatory response initiated by the restoration of blood flow to hypoxic tissue under oxidative stress; prolonged ischemia can cause necrosis. Rather than restoring tissue function, oxidative damage occurs from the induction and accumulation of reactive oxygen species (ROS) from stressed cells. The pathologic effect of HMGB1 in ischemia and I/R is attributed to TLR4/RAGE-dependent ROS production in the solid vital organs.15 HMGB1 is expressed in elevated levels in patients with organ injury or transplantation; hence it is considered a marker of injury.16 Although the pathogenesis of ischemic organ injury is attributed to both TLR4 and RAGE activation, differences in their signaling pathways in different tissues are important.

4.1.1. Liver

In the liver, HMGB1 signals for inflammation via TLR4 and calcium-mediated signaling.1718 The induction of TNF-α and IL-6 and the upregulation of HMGB1 receptors in both ischemic (however more prominently) and nonischemic lobes have been detected.17 In I/R injury, the primary defense mechanism for oxidative damage-induced cellular damage is elevation of manganese superoxide dismutase.19 IL-1R-associated kinase-M-dependent suppression of TLR4 signaling was also found to dampen inflammation in mice when pretreated with HMGB1.20 The release of HMGB1 and macrophage migration inhibitory factor, a coexpressed DAMP, is indicative of the extent of liver damage.21

4.1.2. Heart

RAGE is the prominent signaling pathway in cardiac I/R injury, signaling for the expression of proinflammatory genes via mitogen-activated protein kinase (MAPK), the JAK-STAT signaling family, and others2223; TLR4 is also activated. An array of enzymes and molecules produced in response to a hypoxic insult, such as lactate dehydrogenase, creatine kinase, superoxide dismutase (SOD), and malondialdehyde, can be used as markers to determine the extent of cardiac damage, corresponding to infarct size.2425 The inhibition of HMGB1 by, for example, geranylgeranylacetone (a small molecule inhibitor) therefore lowers the expression of these markers.24 After I/R injury, there is an increase in the release of advanced glycation end-products (AGEs) that also increases HMGB1 expression, causing inflammation.23 The acute damage initiated by ischemia or I/R is also thought to be a possible initiator of chronic cardiac inflammatory diseases such as hypertrophy.26 However, in preventing I/R injury, hypoxic cardiac reoxygenation in an initial reperfusion has been shown to decrease oxidative damage as a result of HMGB1 downregulation.2728 The protective effects also extend to the respiratory system, in the event of cardiopulmonary bypass being used.27

4.1.3. Kidney

Renal ischemic or I/R injury is significant after transplantation, and the expression of HMGB1 and other injurious molecules plays a crucial role in determining the success or rejection of transplantation.293031 The injury is attributed significantly to HMGB1- and TLR4-mediated inflammation that destroys the tubular architecture and induces cell death32; this is mediated at least partly in an alpha-2-adrenoreceptor-dependent manner that suppresses the inflammatory feedforward circuit.33 In acute ischemic injury, there is an increase in TLR4 expression in the proximal tubule endothelial cells near the cortex and outer medulla.34 Renal HMGB1 expression is the key to IL-6 release by infiltrating macrophages that promote inflammatory cytokine production.35 Furthermore, renal endothelial activation of TLR4 induces the expression of NKG2D ligands that renders them susceptible to cytotoxic killing.36

4.1.4. Nervous system

Abundant literature on ischemic and I/R injury in the cerebral and nervous systems suggests that HMGB1 is integral to organ injury. HMGB1 has cytokine-like functions in tissues of the nervous system, and is found in activated microglia and astrocytes, as well as the microvasculature of the infarction core.37 The number of TLRs, especially TLR4, and the signaling strength are proportional to the extent of the damage and the infarct size.383940 Similarly to I/R cardiac injury, hypoxic reperfusion techniques have been used in spinal cord reperfusion, such as the use of hydrogen gas.41 Hemostasis in reperfusion is important to postischemic brain injury as large von Willebrand factor multimers have an HMGB1-associated proinflammatory function that is exacerbated by ADAMTS13–/–.42 HMGB1 release from glial cells is partly mediated by the alpha7 nicotinic acetylcholine receptor (α7nAChR), whose expression directly correlates to the number of apoptotic cells in the ischemic penumbra.43 The inhibition of ischemia-induced apoptosis is also seen by the activation of hemeoxygenase-1 via phosphoinositide 3-kinase/Akt signaling pathways and nuclear factor (erythroid-derived 2)-like 2 (Nrf-2) translocation.44 Not only are the nerves susceptible, but the eye is also subject to HMGB1- and RAGE-mediated retinal damage.45 Furthermore, neuronal zymogen matrix metalloproteinase 9 activation in response to HMGB1 release is another key contributor to tissue damage and the release of neurotoxic ligands.46 Glutamate-induced cytotoxicity is another mechanism of damage following acute ischemic damage that can propagate through neural tissue, forming the ischemic penumbra.47

Acute organ injury from ischemia or I/R injury has been investigated in other organs and for quantitative assaying of cellular damage as well, such as in islet cell preparations for xenotransplantation.48 The different tissue types and expression of HMGB1 in ischemia and I/R organ injury are subject to distinct tissue types and signaling mechanisms.

4.2. Organ injury from chronic and atherosclerotic damage

HMGB1 is also a key contributor mediating organ injury in chronic inflammatory diseases. Recent research has paid close attention to its importance in the pathogenesis of atherosclerosis and other cardiomyopathies, pulmonary diseases such as chronic obstructive pulmonary disease (COPD), and diabetes. The signaling mechanisms of DAMP receptors contribute differently to these diseases.

4.2.1. Atherosclerosis and chronic cardiomyopathies

Atherosclerosis is a chronic cardiovascular disease that arises from plaque formation on the arterial walls that is composed of infiltrated macrophages, oxidized cholesterol, and calcification.4950 HMGB1 alteration of endothelial adhesion, particularly the expression of intercellular adhesion molecule 1, vascular cell adhesion molecule 1, and monocyte chemotactic protein 1, which is crucial in inflammocyte recruitment, in addition to the conventional array of proinflammatory cytokines it induces, is responsible for lesion formation and growth.49 Plaque growth is a complex process that is fueled by inflammation significantly caused by HMGB1; it includes entrapment of low-density lipoprotein in the endothelia, monocyte recruitment, and foam cell formation as a result of monocytic internalization of lipids. The growing plaque is stabilized by the secretion of metalloproteinases, forming a fibrous cap, along with other cytokines, ROS, and growth factors.51 The symptoms then manifest once the plaque ruptures from instability, causing thrombosis.

A key characteristic of progressing atherosclerotic lesions is the lipid or necrotic core, which may contain dying macrophages, where intense levels of HMGB1 expression are found.52 Not only is the elevation of HMGB1 expression found in macrophages, but also in the vascular smooth muscle cells of the tunica intima and adventitia, and in the endothelia of vessels that contain atherosclerotic lesions.5354 RAGE is thought to be the main HMGB1 receptor responsible for chronic inflammation, as its expression is elevated in all cells involved in atherosclerosis,55 and as elevated AGEs are also thought to contribute greatly to the initial endothelial transformation.56 However, the importance of TLR4 signaling must not be neglected, as its attenuation (e.g., by statins) decreases inflammation and plaque growth to a remarkable extent,57 partly by inhibiting IL-6 secretion.58

HMGB1 is also integral to the pathogenesis of other chronic inflammatory-mediated cardiac diseases. Chronic myocarditis is a chronic inflammatory disorder that involves ventricular remodeling, hypertrophy, and fibrosis that may potentially be mediated by HMGB1 in a mainly RAGE-dependent mechanism similar to those mentioned regarding atherosclerosis26; this also includes ventricular remodeling postmyocardial infarction. Furthermore, HMGB1 is also a marker for different levels of fibrosis in chronic hepatitis.59

4.2.2. Chronic pulmonary disease

Chronic inflammation is involved in the pathogenesis of many pulmonary diseases, such as COPD, oxidative stress, protease–antiprotease imbalance and inflammation, the pathogenic triad of COPD whose features include chronic bronchitis and emphysema and have been shown to involve HMGB1.60 Elevated levels of HMGB1 are found in the peripheral airways of patients with COPD, more so in smokers than nonsmokers; the levels of expressed IL-8 and polymorphonuclear elastase, important for inflammocyte recruitment and airway destruction, were also elevated in a similar manner.6162 The deficiency of soluble RAGE to compete with those receptors expressed on pulmonary/inflammatory cells also contributes to the pathogenesis of COPD.63

4.2.3. Diabetes-related diseases

Other chronic diseases to which HMGB1-mediated inflammation may contribute are diabetes-related illnesses. The development of diabetic nephropathy is an inflammatory process, marked by an increase in TLR4 expression on renal tubular cells.64 Like that in the arteries, glomerulosclerosis from tubular fibrosis and injury is caused by the expression of proinflammatory molecules, in particular chemokine (C–C motif) ligand 2 (CCL2), which causes monocyte/macrophage infiltration. In addition, hyperglycemic states can increase TLR2 and TLR4 expression in monocytes.65

RAGE and TLR4 are responsible not only for glomerulosclerosis, but also for diabetic atherosclerosis and cardiovascular diseases.6667 Due to the accumulation of AGEs in diabetic kidney disease, RAGE has been greatly implicated in glomerular, tubular, and vascular dysfunction, especially in the loss of podocyte cells, causing proteinuria.68 HMGB1-dependent RAGE signaling is highly toxic to pancreatic beta islets, resulting in induction of apoptosis and inhibition of insulin secretion. This ultimately results in the perpetuation and exacerbation of type 2 diabetes mellitus.69 Moreover, HMGB1 has been found to be involved in diabetic allodynia by causing astrocytotic activation.70

4.2.4. Chronic kidney diseases

Other chronic kidney diseases that are not related to diabetes are also subject to HMGB1-mediated organ injury. It has been shown that attenuating HMGB1 could effectively treat chronic kidney disease with sepsis in mice, but not sepsis alone, suggesting that it is an important mediator in chronic kidney disease.71 HMGB1-dependent RAGE activation has been shown to be integral to amplifying vascular damage in chronic kidney diseases.68 It was also correlated with indicators of inflammation and malnutrition in end-stage renal patients,72 and correlated directly with renal function atherosclerosis in uremia.73 Chronic damage by HMGB1 includes fibrosis, structural damage, sclerosis, and granulomatous inflammatory lesions that are caused by DAMP receptor signaling.74 TLR2 has been especially attributed to tubular renal matrix accumulation and injury, in addition to TLR4, TLR9, and RAGE signaling.75

4.2.5. Neuroinflammation and neurodegenerative diseases

Evidence from research has supported the role of HMGB1 in neurodegenerative diseases via activation of RAGE in Alzheimer's disease, and TLR4 in multiple sclerosis. Controlled neuroinflammation is important to normal neurologic function, as inflammatory cytokines such as IFN-γ are important in decreasing neuronal oxidative stress by inducing gamma-interferon inducible lysosomal thiol reductase (GILT) and superoxide dismutase 2 (SOD2) activity.76 However, deregulation of neuroinflammation by HMGB1 increases inflammation to toxic levels, and is a cause of pathology.77 Neuronal dysfunction and cognitive deficits are hallmarks of neuroinflammation that are significantly caused by TLR4-dependent TNF-α secretion by activated microglia.77 Hyperexcitability is another hallmark of neuroinflammation that is directly linked to apoptosis. TLR-mediated signaling by IL-1β, a cytokine induced by HMGB1, has been shown to be involved in increasing the hyperexcitation of epileptogenic tissue through modulating post-translational changes in voltage-gated ion channels and transcription of genes affecting neurotransmission and neuroplasticity.78 Mechanistic studies have shown that microglial Mac1 receptors are also able to interact with HMGB1 to express NFκB and NADPH oxidase, producing inflammatory and neurotoxic factors.79 TLR and Mac1 are implicated greatly in the neurodegenerative processes underlying multiple sclerosis, Parkinson's disease, and seizures.

RAGE has also been subjected to vast research in the pathogenesis of Alzheimer's disease as it is able to interact with the neurotoxic beta-amyloid peptide. The ability of HMGB1 and other RAGE ligands to activate and upregulate the expression of RAGE makes it a key contributor to exacerbations of neurotoxicity in Alzheimer's disease.80 RAGE is responsible for transcytosis of beta-amyloid across the blood–brain barrier, and inflammatory signaling resulting in the expression of NFκB and neurotoxic cytokines.8182 Its activation has also been implicated as a cause of beta-amyloid aggregation and plaque formation, inducing apoptosis by oxidative stress, in addition to the induction of the proinflammatory milieu in microglia.83 The ability of HMGB1 to interact with TLRs, RAGE, and Mac1 in a neurologic setting makes it a key mediator of neuroinflammation and neuronal injury.

4.2.6. Mechanical stress and systemic inflammation

Mechanical or shear/traumatic stress is the basis of organ damage from accidents or from surgery. In clinical settings where the patient is subjected to significant physical trauma, the systemic inflammatory response syndrome is often present, which can lead to multiple organ failure/damage; the expression of IL-6 and TNF-α has been shown to be induced by HMGB1, followed by increased RAGE expression at the site of injury.84 The risk for developing sepsis from multiple organ dysfunction can be assessed by single-nucleotide polymorphisms present in the HMGB1 gene.85 The plasma levels of HMGB1 are correlated to mast cell activation, but it is still unknown whether this is due to less organ damage in mast cell-deficient mice, or to regulation of HMGB1 release by mast cells.86

In surgical procedures of the liver that involve compression or clamping, organ injury can arise not only from ischemic damage, but from trauma as well. Neutrophil elastase-mediated damage is directly linked with the chemotactic properties of HMGB1 signaling.87 Experimental data suggest that mechanical stretch in the alveolar epithelial cells stimulates the expression of HMGB1 and the activity of MKK6, an MAPK known to regulate apoptosis.8889 The predictive value of HMGB1 levels for lung injury, sepsis, and multiple organ injury is also shown in postoperative thoracic esophagectomy patients, as those with complications had higher postoperative serum HMGB1 levels.90 The pathogenesis of injury of different organs arising from systemic inflammatory responses seems to be due to the feedforward hyperstimulation of TLR4- and RAGE-mediated mechanisms that were discussed previously for HMGB1.

4.3. Acute toxicity

Other forms of acute organ injury that are not related to ischemic damage are also influenced by HMGB1. Allograft rejection and drug/nondrug-related toxicity are two major areas that have been reported frequently in recent literature.

4.3.1. Allograft rejection

The role of HMGB1 as an alarmin in allograft rejection and the success of organ transplantation has been investigated. The secretion of IL-6 from dendritic cells that induces the expression of IL-17 in alloreactive T-cells has been shown in early-phase acute cardiac allograft rejection, usually followed by T helper type 1 cell responses that produce IFN-γ, provoking graft destruction.91 Nonchronic fibrosing conditions are the result of TLR2/4 signaling in both chronic and acute transplant settings, directly and significantly affecting renal allograft function.92 It has also been shown that chronic vasculopathies result from HMGB1-dependent RAGE signaling that produces vascular endothelial growth factor.93 Both HMGB1 and its cytokine IL-1β are responsible for orchestrating the strength of rejection reactions.94 Being involved in the exacerbation of allograft rejections of many organs, such as the liver, kidney, heart, islet cells, and vessels, HMGB1 can be used as a marker for transplant success before and after organ harvest.309596

4.3.2. Acute toxicity and lesions

Acute toxicity has also been reported to damage organs through HMGB1 release. In acetaminophen-induced hepatotoxicity, HMGB1 is a marker for apoptotic and necrotic cell death that is used to assess survival.97 Cytoplasmic translocation of HMGB1 occurs in stressed hepatocytes, and HMGB1 is also released from dead cells.98 More importantly, the extent of acute damage is determined by hepatocyte expression of CXCR3 crucial for immunocyte infiltration.99 Although recent research has focused on the liver in acute toxicity, acute inflammatory lesions generally signal via TLR4, implying that HMGB1 is a key player in the disease process.100

5. HMGB1 and the prevention of organ damage

Besides conventional inhibition of HMGB1 or its receptors by antibodies, much of the current literature tries to elucidate different mechanisms of HMGB1 release and signaling, revealing different methods of inhibiting its release, or the damage that is caused. These methods include splenectomies, small-molecule inhibitors, anti-HMGB1 antibodies, and competitive agents that decrease DAMP receptor activation. HMGB1 preconditioning has been shown to decrease the inflammatory response by activating the natural signal-dampening mechanism.20 Techniques of reperfusion, such as hypoxic reoxygenation and thermoregulation, have also been shown to decrease inflammation.1728

As for xenobiotics, statins and ethyl pyruvate inhibit the cytokine response and decrease HMGB1 release, respectively.58101 Small inhibitors like glycyrrhizin, geranylgeranylacetone and dexmedetomidine, neutrophil elastase inhibitors, and lysosomal thiol reductase inhibitors, have recently been studied and shown to inhibit HMGB1-mediated organ injury.33437687102 The current discovery of receptors involved in HMGB1-mediated organ injury has also led to alpha-2-adrenergic receptor inhibition by dexmedetomidine and electroacupuncture for α7nAChR attenuation to decrease inflammation.3343 Furthermore, using soluble RAGE to compete for HMGB1-dependent RAGE activation has also been shown to decrease organ injury.63 The study of HMGB1 and its signaling pathways continues to increase the possibilities in drug design and therapy to prevent and/or treat organ injury.

6. Conclusions

HMGB1 is a key mediator of inflammation that results in both acute and chronic organ injury. Although vital to a healthy immune system and initiating healing, its multireceptor interaction with TLRs and RAGE, and its feedforward mechanistic nature, is the key to its role in the pathogenesis of diseases, resulting in damage to the vital organs.


References

1
H. Yanai, T. Ban, Z. Wang, M.K. Choi, T. Kawamura, H. Negishi, et al.
HMGB proteins function as universal sentinels for nucleic-acid-mediated innate immune responses
Nature, 462 (2009), pp. 99-103
2
H. Yanai, S. Chiba, T. Ban, Y. Nakaima, T. Onoe, K. Honda, et al.
Suppression of immune responses by nonimmunogenic oligodeoxynucleotides with high affinity for high-mobility group box proteins (HMGBs)
Proc Natl Acad Sci U S A, 108 (2011), pp. 11542-11547
3
K.M. Livesey, R. Kang, P. Vernon, W. Buchser, P. Loughran, S.C. Watkins, et al.
p53/HMGB1 complexes regulate autophagy and apoptosis
Cancer Res, 72 (2012), pp. 1996-2005
4
Y. Miyake, S. Yamasaki
Sensing necrotic cells
Adv Exp Med Biol, 738 (2012), pp. 144-152
5
L. Vande Walle, T.D. Kanneganti, M. Lamkanfi
HMGB1 release by inflammasomes
Virulence, 2 (2011), pp. 162-165
CrossRef  
6
J.O. Thomas, K. Stott
H1 and HMGB1: Modulators of chromatin structure
Biochem Soc Trans, 40 (2012), pp. 341-346
7
R. Chakraborty, K.H. Bhatt, A. Sodhi
Ultraviolet B induces high mobility group box 1 release from mouse peritoneal macrophages in vitro via caspase-1 mediated secretion pathway
Immunobiology, 218 (2013), pp. 135-144
8
S. Shiozawa, K. Tsumiyama
Pathogenesis of rheumatoid arthritis and c-Fos/AP-1
Cell Cycle, 8 (2009), pp. 1539-1543
9
K. Hayakawa, J. Qiu, E.H. Lo
Biphasic actions of HMGB1 signaling in inflammation and recovery after stroke
Ann N Y Acad Sci, 1207 (2010), pp. 50-57
10
U. Andersson, K.J. Tracey
HMGB1 is a therapeutic target for sterile inflammation and infection
Annu Rev Immunol, 29 (2011), pp. 139-162
11
H.E. Harris, U. Andersson, D.S. Pisetsky
HMGB1: A multifunctional alarmin driving autoimmune and inflammatory disease
Nat Rev Rheumatol, 8 (2012), pp. 195-202
12
M. Schiraldi, A. Raucci, L.M. Munoz, E. Livoti, B. Celona, E. Venereau, et al.
HMGB1 promotes recruitment of inflammatory cells to damaged tissues by forming a complex with CXCL12 and signaling via CXCR4
J Exp Med, 209 (2012), pp. 551-563
13
J.R. van Beijnum, W.A. Buurman, A.W. Griffioen
Convergence and amplification of toll-like receptor (TLR) and receptor for advanced glycation end products (RAGE) signaling pathways via high mobility group B1 (HMGB1)
Angiogenesis, 11 (2008), pp. 91-99
14
G.Y. Chen, J. Tang, P. Zheng, Y. Liu
CD24 and siglec-10 selectively repress tissue damage-induced immune responses
Science, 323 (2009), pp. 1722-1725
15
D.J. Kaczorowski, A. Tsung, T.R. Billiar
Innate immune mechanisms in ischemia/reperfusion
Front Biosci (Elite Ed), 1 (2009), pp. 91-98
16
D. Tang, R. Kang, H.J. Zeh 3rd, M.T. Lotze
High-mobility group box 1, oxidative stress, and disease
Antioxid Redox Signal, 14 (2011), pp. 1315-1335
17
A. Liu, O. Dirsch, H. Fang, J. Sun, H. Jin, W. Dong, et al.
HMGB1 in ischemic and non-ischemic liver after selective warm ischemia/reperfusion in rat
Histochem Cell Biol, 135 (2011), pp. 443-452
18
A. Tsung, J.R. Klune, X. Zhang, G. Jeyabalan, Z. Cao, X. Peng, et al.
HMGB1 release induced by liver ischemia involves toll-like receptor 4 dependent reactive oxygen species production and calcium-mediated signaling
J Exp Med, 204 (2007), pp. 2913-2923
19
M. Pardo, N. Budick-Harmelin, B. Tirosh, O. Tirosh
Antioxidant defense in hepatic ischemia-reperfusion injury is regulated by damage-associated molecular pattern signal molecules
Free Radic Biol Med, 45 (2008), pp. 1073-1083
20
K. Izuishi, A. Tsung, G. Jeyabalan, N.D. Critchlow, J. Li, K.J. Tracey, et al.
Cutting edge: High-mobility group box 1 preconditioning protects against liver ischemia-reperfusion injury
J Immunol, 176 (2006), pp. 7154-7158
21
A. Liu, H. Jin, O. Dirsch, M. Deng, H. Huang, M. Brocker-Preuss, et al.
Release of danger signals during ischemic storage of the liver: A potential marker of organ damage?
Mediators Inflamm, 2010 (2010), p. 436145
Article  
22
R. Ramasamy, A.M. Schmidt
Receptor for advanced glycation end products (RAGE) and implications for the pathophysiology of heart failure
Curr Heart Fail Rep, 9 (2012), pp. 107-116
23
S.F. Yan, R. Ramasamy, A.M. Schmidt
The receptor for advanced glycation endproducts (RAGE) and cardiovascular disease
Expert Rev Mol Med, 11 (2009) e9
Article  
24
N. Wang, X. Min, D. Li, P. He, L. Zhao
Geranylgeranylacetone protects against myocardial ischemia and reperfusion injury by inhibiting high-mobility group box 1 protein in rats
Mol Med Rep, 5 (2012), pp. 521-524
25
S. Gratia, L. Kay, L. Potenza, A. Seffouh, V. Novel-Chate, C. Schnebelen, et al.
Inhibition of AMPK signalling by doxorubicin: At the crossroads of the cardiac responses to energetic, oxidative, and genotoxic stress
Cardiovasc Res, 95 (2012), pp. 290-299
26
H.C. Volz, Z. Kaya, H.A. Katus, M. Andrassy
The role of HMGB1/RAGE in inflammatory cardiomyopathy
Semin Thromb Hemost, 36 (2010), pp. 185-194
27
J. Rong, S. Ye, Z.K. Wu, G.X. Chen, M.Y. Liang, H. Liu, et al.
Controlled oxygen reperfusion protects the lung against early ischemia-reperfusion injury in cardiopulmonary bypasses by downregulating high mobility group box 1
Exp Lung Res, 38 (2012), pp. 183-191
28
U. Abdel-Rahman, P. Risteski, K. Tizi, S. Kerscher, S. Behjati, K. Zwicker, et al.
Hypoxic reoxygenation during initial reperfusion attenuates cardiac dysfunction and limits ischemia-reperfusion injury after cardioplegic arrest in a porcine model
J Thorac Cardiovasc Surg, 137 (2009), pp. 978-982
29
C.Y. Lu, P.D. Winterberg, J. Chen, J.R. Hartono
Acute kidney injury: A conspiracy of toll-like receptor 4 on endothelia, leukocytes, and tubules
Pediatr Nephrol, 27 (2012), pp. 1847-1854
30
D. Kaminska, K. Koscielska-Kasprzak, D. Drulis-Fajdasz, A. Halon, W. Polak, P. Chudoba, et al.
Kidney ischemic injury genes expressed after donor brain death are predictive for the outcome of kidney transplantation
Transplant Proc, 43 (2011), pp. 2891-2894
31
H. Wu, J. Ma, P. Wang, T.M. Corpuz, U. Panchapakesan, K.R. Wyburn, et al.
HMGB1 contributes to kidney ischemia reperfusion injury
J Am Soc Nephrol, 21 (2010), pp. 1878-1890
32
T.A. Sutton, P.C. Dagher
Fueling the fire in acute kidney injury: Endothelial cells collect their toll
Kidney Int, 79 (2011), pp. 267-269
33
J. Gu, P. Sun, H. Zhao, H.R. Watts, R.D. Sanders, N. Terrando, et al.
Dexmedetomidine provides renoprotection against ischemia-reperfusion injury in mice
Crit Care, 15 (2011) R153
Article  
34
J. Chen, R. John, J.A. Richardson, J.M. Shelton, X.J. Zhou, Y. Wang, et al.
Toll-like receptor 4 regulates early endothelial activation during ischemic acute kidney injury
Kidney Int, 79 (2011), pp. 288-299
35
J. Chen, J.R. Hartono, R. John, M. Bennett, X.J. Zhou, Y. Wang, et al.
Early interleukin 6 production by leukocytes during ischemic acute kidney injury is regulated by TLR4
Kidney Int, 80 (2011), pp. 504-515
36
G.E. Chen, H. Wu, J. Ma, S.J. Chadban, A. Sharland
Toll-like receptor 4 engagement contributes to expression of NKG2D ligands by renal tubular epithelial cells
Nephrol Dial Transplant, 26 (2011), pp. 3873-3881
37
J.B. Kim, C.M. Lim, Y.M. Yu, J.K. Lee
Induction and subcellular localization of high-mobility group box-1 (HMGB1) in the postischemic rat brain
J Neurosci Res, 86 (2008), pp. 1125-1131
38
Y.C. Wang, S. Lin, Q.W. Yang
Toll-like receptors in cerebral ischemic inflammatory injury
J Neuroinflammation, 8 (2011), p. 134
39
Q.W. Yang, F.L. Lu, Y. Zhou, L. Wang, Q. Zhong, S. Lin, et al.
HMBG1 mediates ischemia-reperfusion injury by TRIF-adaptor independent toll-like receptor 4 signaling
J Cereb Blood Flow Metab, 31 (2011), pp. 593-605
40
Q.W. Yang, J. Xiang, Y. Zhou, Q. Zhong, J.C. Li
Targeting HMGB1/TLR4 signaling as a novel approach to treatment of cerebral ischemia
Front Biosci (Schol Ed), 2 (2010), pp. 1081-1091
41
Y. Huang, K. Xie, J. Li, N. Xu, G. Gong, G. Wang, et al.
Beneficial effects of hydrogen gas against spinal cord ischemia-reperfusion injury in rabbits
Brain Res, 1378 (2011), pp. 125-136
42
M. Fujioka, T. Nakano, K. Hayakawa, K. Irie, Y. Akitake, Y. Sakamoto, et al.
ADAMTS13 gene deletion enhances plasma high-mobility group box1 elevation and neuroinflammation in brain ischemia-reperfusion injury
Neurol Sci, 33 (2012), pp. 1107-1115
43
Q. Wang, F. Wang, X. Li, Q. Yang, X. Li, N. Xu, et al.
Electroacupuncture pretreatment attenuates cerebral ischemic injury through alpha7 nicotinic acetylcholine receptor-mediated inhibition of high-mobility group box 1 release in rats
J Neuroinflammation, 9 (2012), p. 24
44
Y.M. Ha, M.Y. Kim, M.K. Park, Y.S. Lee, Y.M. Kim, H.J. Kim, et al.
Higenamine reduces HMGB1 during hypoxia-induced brain injury by induction of heme oxygenase-1 through PI3K/Akt/Nrf-2 signal pathways
Apoptosis, 17 (2012), pp. 463-474
45
G. Dvoriantchikova, E. Hernandez, J. Grant, A.R. Santos, H. Yang, D. Ivanov
The high-mobility group box-1 nuclear factor mediates retinal injury after ischemia reperfusion
Invest Ophthalmol Vis Sci, 52 (2011), pp. 7187-7194
46
J. Qiu, J. Xu, Y. Zheng, Y. Wei, X. Zhu, E.H. Lo, et al.
High-mobility group box 1 promotes metalloproteinase-9 upregulation through toll-like receptor 4 after cerebral ischemia
Stroke, 41 (2010), pp. 2077-2082
47
R. Yue, X. Yuan, X. Liu, J. Zhang, P. Jiang, C. He, et al.
Cynandione A mitigates ischemic injuries in rats with cerebral ischemia
J Neurochem, 121 (2012), pp. 451-464
48
T. Itoh, S. Iwahashi, M. Shimoda, D. Chujo, M. Takita, J.A. SoRelle, et al.
High-mobility group box 1 expressions in hypoxia-induced damaged mouse islets
Transplant Proc, 43 (2011), pp. 3156-3160
49
A.W. de Souza, J. Westra, P.C. Limburg, M. Bijl, C.G. Kallenberg
HMGB1 in vascular diseases: Its role in vascular inflammation and atherosclerosis
Autoimmun Rev, 11 (2012), pp. 909-917
50
S. Morris-Rosenfeld, E. Blessing, M.R. Preusch, C. Albrecht, A. Bierhaus, M. Andrassy, et al.
Deletion of bone marrow-derived receptor for advanced glycation end products inhibits atherosclerotic plaque progression
Eur J Clin Invest, 41 (2011), pp. 1164-1171
51
M.J. Meens, A. Pfenniger, B.R. Kwak
Risky communication in atherosclerosis and thrombus formation
Swiss Med Wkly, 142 (2012) w13553
Article  
52
N. Kalinina, A. Agrotis, Y. Antropova, G. DiVitto, P. Kanellakis, G. Kostolias, et al
Increased expression of the DNA-binding cytokine HMGB1 in human atherosclerotic lesions: Role of activated macrophages and cytokines
Arterioscler Thromb Vasc Biol, 24 (2004), pp. 2320-2325
53
S. Park, S.J. Yoon, H.J. Tae, C.Y. Shim
RAGE and cardiovascular disease
Front Biosci, 16 (2011), pp. 486-497
54
K. Inoue, K. Kawahara, K.K. Biswas, K. Ando, K. Mitsudo, M. Nobuyoshi, et al.
HMGB1 expression by activated vascular smooth muscle cells in advanced human atherosclerosis plaques
Cardiovasc Pathol, 16 (2007), pp. 136-143
55
S. Del Turco, G. Basta
An update on advanced glycation endproducts and atherosclerosis
Biofactors, 38 (2012), pp. 266-274
56
J.A. Mosquera
Role of the receptor for advanced glycation end products (RAGE) in inflammation
Invest Clin, 51 (2010), pp. 257-268
57
J. Yang, C. Huang, J. Yang, H. Jiang, J. Ding
Statins attenuate high mobility group box-1 protein induced vascular endothelial activation: A key role for TLR4/NF-kappaB signaling pathway
Mol Cell Biochem, 345 (2010), pp. 189-195
58
H. Loppnow, L. Zhang, M. Buerke, M. Lautenschlager, L. Chen, A. Frister, et al.
Statins potently reduce the cytokine-mediated IL-6 release in SMC/MNC cocultures
J Cell Mol Med, 15 (2011), pp. 994-1004
59
A. Albayrak, M.H. Uyanik, S. Cerrah, S. Altas, H. Dursun, M. Demir, et al.
Is HMGB1 a new indirect marker for revealing fibrosis in chronic hepatitis and a new therapeutic target in treatment?
Viral Immunol, 23 (2010), pp. 633-638
60
B.M. Fischer, E. Pavlisko, J.A. Voynow
Pathogenic triad in COPD: Oxidative stress, protease-antiprotease imbalance, and inflammation
Int J Chron Obstruct Pulmon Dis, 6 (2011), pp. 413-421
61
H. Kanazawa, Y. Tochino, K. Asai, Y. Ichimaru, T. Watanabe, K. Hirata
Validity of HMGB1 measurement in epithelial lining fluid in patients with COPD
Eur J Clin Invest, 42 (2012), pp. 419-426
62
C. Hou, H. Zhao, L. Liu, W. Li, X. Zhou, Y. Lv, et al.
High mobility group protein B1 (HMGB1) in asthma: Comparison of patients with chronic obstructive pulmonary disease and healthy controls
Mol Med, 17 (2011), pp. 807-815
63
M.B. Sukkar, L.G. Wood, M. Tooze, J.L. Simpson, V.M. McDonald, P.G. Gibson, et al.
Soluble RAGE is deficient in neutrophilic asthma and COPD
Eur Respir J, 39 (2012), pp. 721-729
64
M. Lin, W.H. Yiu, H.J. Wu, L.Y. Chan, J.C. Leung, W.S. Au, et al.
Toll-like receptor 4 promotes tubular inflammation in diabetic nephropathy
J Am Soc Nephrol, 23 (2012), pp. 86-102
65
M.R. Dasu, S. Devaraj, L. Zhao, D.H. Hwang, I. Jialal
High glucose induces toll-like receptor expression in human monocytes: Mechanism of activation
Diabetes, 57 (2008), pp. 3090-3098
66
M. Li, L. Song, X. Gao, W. Chang, X. Qin
Toll-like receptor 4 on islet beta cells senses expression changes in high-mobility group box 1 and contributes to the initiation of type 1 diabetes
Exp Mol Med, 44 (2012), pp. 260-267
67
R. Ramasamy, S.F. Yan, A.M. Schmidt
Receptor for AGE (RAGE): Signaling mechanisms in the pathogenesis of diabetes and its complications
Ann N Y Acad Sci, 1243 (2011), pp. 88-102
68
V. D'Agati, A.M. Schmidt
RAGE and the pathogenesis of chronic kidney disease
Nat Rev Nephrol, 6 (2010), pp. 352-360
69
Y. Zhu, T. Shu, Y. Lin, H. Wang, J. Yang, Y. Shi, et al.
Inhibition of the receptor for advanced glycation endproducts (RAGE) protects pancreatic beta-cells
Biochem Biophys Res Commun, 404 (2011), pp. 159-165
70
P.C. Ren, Y. Zhang, X.D. Zhang, L.J. An, H.G. Lv, J. He, et al.
High-mobility group box 1 contributes to mechanical allodynia and spinal astrocytic activation in a mouse model of type 2 diabetes
Brain Res Bull, 88 (2012), pp. 332-337
71
A. Leelahavanichkul, Y. Huang, X. Hu, H. Zhou, T. Tsuji, R. Chen, et al.
Chronic kidney disease worsens sepsis and sepsis-induced acute kidney injury by releasing high mobility group box protein-1
Kidney Int, 80 (2011), pp. 1198-1211
72
N. Zhu, W. Yuan, Y. Zhou, J. Liu, J. Bao, J. Hao, et al.
High mobility group box protein-1 correlates with microinflammatory state and nutritional status in continuous ambulatory peritoneal dialysis patients
J Artif Organs, 14 (2011), pp. 125-132
73
A. Bruchfeld, A.R. Qureshi, B. Lindholm, P. Barany, L. Yang, P. Stenvinkel, et al.
High mobility group box protein-1 correlates with renal function in chronic kidney disease (CKD)
Mol Med, 14 (2008), pp. 109-115
74
Y. Oyama, T. Hashiguchi, N. Taniguchi, S. Tancharoen, T. Uchimura, K.K. Biswas, et al.
High-mobility group box-1 protein promotes granulomatous nephritis in adenine-induced nephropathy
Lab Invest, 90 (2010), pp. 853-866
75
J.C. Leemans, L.M. Butter, W.P. Pulskens, G.J. Teske, N. Claessen, T. van der Poll, et al.
The role of toll-like receptor 2 in inflammation and fibrosis during progressive renal injury
PLoS One, 4 (2009) e5704
Article  
76
H.S. Chiang, M. Maric
Lysosomal thiol reductase negatively regulates autophagy by altering glutathione synthesis and oxidation
Free Radic Biol Med, 51 (2011), pp. 688-699
77
K. Belarbi, T. Jopson, D. Tweedie, C. Arellano, W. Luo, N.H. Greig, et al.
TNF-alpha protein synthesis inhibitor restores neuronal function and reverses cognitive deficits induced by chronic neuroinflammation
J Neuroinflammation, 9 (2012), p. 23
Article  
78
A. Vezzani, M. Maroso, S. Balosso, M.A. Sanchez, T. Bartfai
IL-1 receptor/Toll-like receptor signaling in infection, inflammation, stress and neurodegeneration couples hyperexcitability and seizures
Brain Behav Immun, 25 (2011), pp. 1281-1289
79
H.M. Gao, H. Zhou, F. Zhang, B.C. Wilson, W. Kam, J.S. Hong
HMGB1 acts on microglia Mac1 to mediate chronic neuroinflammation that drives progressive neurodegeneration
J Neurosci, 31 (2011), pp. 1081-1092
80
E. Leclerc, E. Sturchler, S.W. Vetter
The S100B/RAGE axis in alzheimer's disease
Cardiovasc Psychiatry Neurol, 2010 (2010), p. 539581
Article  
81
S.F. Yan, S.D. Yan, R. Ramasamy, A.M. Schmidt
Tempering the wrath of RAGE: An emerging therapeutic strategy against diabetic complications, neurodegeneration, and inflammation
Ann Med, 41 (2009), pp. 408-422
82
E. Leclerc, E. Sturchler, S.W. Vetter, C.W. Heizmann
Crosstalk between calcium, amyloid beta and the receptor for advanced glycation endproducts in alzheimer's disease
Rev Neurosci, 20 (2009), pp. 95-110
83
S.H. Han, Y.H. Kim, I.R.A.G.E. Mook-Jung
The beneficial and deleterious effects by diverse mechanisms of actions
Mol Cells, 31 (2011), pp. 91-97
84
J. Shimazaki, N. Matsumoto, H. Ogura, T. Muroya, Y. Kuwagata, J. Nakagawa, et al.
Systemic involvement of high-mobility group box 1 protein and therapeutic effect of anti-high-mobility group box 1 protein antibody in a rat model of crush injury
Shock, 37 (2012), pp. 634-638
85
L. Zeng, A.Q. Zhang, W. Gu, K.H. Chen, D.P. Jiang, L.Y. Zhang, et al.
Clinical relevance of single nucleotide polymorphisms of the high mobility group box 1 protein gene in patients with major trauma in southwest china
Surgery, 151 (2012), pp. 427-436
86
C. Cai, Z. Cao, P.A. Loughran, S. Kim, S. Darwiche, S. Korff, et al.
Mast cells play a critical role in the systemic inflammatory response and end-organ injury resulting from trauma
J Am Coll Surg, 213 (2011), pp. 604-615
87
S. Tsujii, T. Okabayashi, M. Shiga, Y. Takezaki, T. Sugimoto, M. Kobayashi, et al.
The effect of the neutrophil elastase inhibitor sivelestat on early injury after liver resection
World J Surg, 36 (2012), pp. 1122-1127
88
N. Ding, F. Wang, Y. Han, H. Xiao, L. Xu, S. She
Mitogen-activated protein kinase kinase 6 mediates mechanical stretch-induced high-mobility group box 1 protein expression in pulmonary alveolar epithelial cells
J Trauma Acute Care Surg, 72 (2012), pp. 162-168
89
A. Matsuzawa, K. Saegusa, T. Noguchi, C. Sadamitsu, H. Nishitoh, S. Nagai, et al.
ROS-dependent activation of the TRAF6-ASK1-p38 pathway is selectively required for TLR4-mediated innate immunity
Nat Immunol, 6 (2005), pp. 587-592
90
K. Suda, Y. Kitagawa, S. Ozawa, T. Miyasho, M. Okamoto, Y. Saikawa, et al.
Neutrophil elastase inhibitor improves postoperative clinical courses after thoracic esophagectomy
Dis Esophagus, 20 (2007), pp. 478-486
91
L. Duan, C.Y. Wang, J. Chen, Q. Gong, P. Zhu, F. Zheng, et al.
High-mobility group box 1 promotes early acute allograft rejection by enhancing IL-6-dependent Th17 alloreactive response
Lab Invest, 91 (2011), pp. 43-53
92
S. Wang, C. Schmaderer, E. Kiss, C. Schmidt, M. Bonrouhi, S. Porubsky, et al.
Recipient toll-like receptors contribute to chronic graft dysfunction by both MyD88- and TRIF-dependent signaling
Dis Model Mech, 3 (2010), pp. 92-103
93
W. Wei, M. Chen, Y. Zhu, J. Wang, P. Zhu, Y. Li, et al.
Down–regulation of vascular HMGB1 and RAGE expression by n–3 polyunsaturated fatty acids is accompanied by amelioration of chronic vasculopathy of small bowel allografts
J Nutr Biochem, 23 (2012), pp. 1333-1340
94
D.A. Rao, J.S. Pober
Endothelial injury, alarmins, and allograft rejection
Crit Rev Immunol, 28 (2008), pp. 229-248
95
Q. Gong, H. Zhang, J.H. Li, L.H. Duan, S. Zhong, X.L. Kong, et al.
High–mobility group box 1 exacerbates concanavalin A–induced hepatic injury in mice
J Mol Med (Berl), 88 (2010), pp. 1289-1298
96
S. Zhang, J. Zhong, P. Yang, F. Gong, C.Y. Wang
HMGB1, an innate alarmin, in the pathogenesis of type 1 diabetes
Int J Clin Exp Pathol, 3 (2009), pp. 24-38
97
D.G. Craig, P. Lee, E.A. Pryde, G.S. Masterton, P.C. Hayes, K.J. Simpson
Circulating apoptotic and necrotic cell death markers in patients with acute liver injury
Liver Int, 31 (2011), pp. 1127-1136
98
R.R. Zhou, S.S. Zhao, M.X. Zou, P. Zhang, B.X. Zhang, X.H. Dai, et al.
HMGB1 cytoplasmic translocation in patients with acute liver failure
BMC Gastroenterol, 11 (2011), p. 21
99
M.M. Zaldivar, M.L. Berres, H. Sahin, A. Nellen, D. Heinrichs, P. Schmitz, et al.
The chemokine receptor CXCR3 limits injury after acute toxic liver damage
Lab Invest, 92 (2012), pp. 724-734
100
H. Hirano, T. Yoshioka, S. Yunoue, S. Fujio, H. Yonezawa, T. Niiro, et al.
TLR4, IL–6, IL–18, MyD88 and HMGB1 are highly expressed in intracranial inflammatory lesions and the IgG4/IgG ratio correlates with TLR4 and IL–6
Neuropathology, 32 (2012), pp. 628-637
101
S.H. Dave, J.S. Tilstra, K. Matsuoka, F. Li, R.A. DeMarco, D. Beer–Stolz, et al.
Ethyl pyruvate decreases HMGB1 release and ameliorates murine colitis
J Leukoc Biol, 86 (2009), pp. 633-643
102
G. Gong, L.B. Yuan, L. Hu, W. Wu, L. Yin, J.L. Hou, et al.
Glycyrrhizin attenuates rat ischemic spinal cord injury by suppressing inflammatory cytokines and HMGB1
Acta Pharmacol Sin, 33 (2012), pp. 11-18

References

Close