Loading

Annals of Vascular Medicine and Research

HMGB1 as a Therapeutic Target to Alleviate Endothelial Dysfunction in Severe Systemic Inflammation

Review Article | Open Access | Volume 10 | Issue 3

  • 1. Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research, USA
  • 2. Institute of Health and Sport, Victoria University, Australia
  • 3. Department of Women’s and Children’s Health, Karolinska Institute, Sweden
+ Show More - Show Less
Corresponding Authors
Ulf Andersson, Department of Women’s and Children’s Health, Karolinska Institute, Stockholm 17176, Sweden
Abstract

Excessive endothelial activation causing microvascular dysfunction is key in the pathogenesis of severe systemic inflammation. The ubiquitous nuclear protein high-mobility group box-1 (HMGB1) is extracellularly released as an alarmin and elicits potent proinflammatory effects on endothelial cells after sterile or infectious tissue injury. These effects include increased release of cytokines, chemokines, and factors promoting coagulation and fibrinolytic activities, and luminal endothelial expression of adhesion molecules. HMGB1 is actively discharged from innate immune cells, endothelial cells, and sensory-motor nerves innervating vascular compartments. Discrete HMGB1 redox isoforms signal via toll-like receptor 4 or receptor for advanced glycation end-products (RAGE) to induce inflammation. Furthermore, extracellular HMGB1 avidly complex-binds other extracellular proinflammatory molecules and these complexes are endocytosed via RAGE to the endolysosomal system in many cells, such as endothelial cells. Increased intralysosomal HMGB1 levels disrupt the lysosomal membrane allowing HMGB1-transported co-molecules access to cognate cytoplasmic proinflammatory receptors and inflammasome systems. The subsequent intracellular activation causes endothelial cell pyroptosis, liberating potent procoagulant and proinflammatory molecules. Therapies based on HMGB1-binding antagonists have yielded mixed results in preclinical studies of systemic inflammation. Unsuccessful results were possibly caused by steric hindrance for HMGB1- binding antagonists due to HMGB1 complex formation. An alternative therapeutic strategy is to inhibit HMGB1 release, a process requiring acetylation of nuclear HMGB1 enabling cytoplasmic export. Increased histone deacetylase activity reduces HMGB1 release and is promoted by acetylcholine signaling via alpha 7 nicotinic acetylcholine receptors present on endothelial cells and sensory neurons. Invasive and non-invasive electrical vagus nerve stimulation activates and mobilizes acetylcholine-secreting anti-inflammatory T lymphocytes to the microvascular compartment. This review focuses on determining the role that the innate immune system and sensory nerves have on driving mechanisms of endothelial dysfunction via release of pathogenic extracellular HMGB1.

Keywords

Endothelium,  Endothelium Dysfunction,  High-Mobility Group Box-1, Inflammasome, Inflammation, Microcirculation, Sensory Neurons, Sepsis.

CITATION

Yang H, Gadanec LK, Zulli A, Andersson U. (2023) HMGB1 as a Therapeutic Target to Alleviate Endothelial Dysfunction in Severe Systemic Inflammation. Ann Vasc Med Res 10(3): 1164.

INTRODUCTION

The endothelium constitutes a physiologically dynamic single cell interface, which lines blood and lymphatic vessels and provides communication between circulating blood and tissues to maintain homeostasis by secreting various vasoactive molecules in response to physical and chemical stimuli and signals [1]. The endothelium promotes homeostasis by (i) tightly controlling vascular tone, permeability and remodeling; (ii) regulating blood fluidity, angiogenesis, and thrombosis; (iii) driving anti-inflammatory, -oxidative and -proliferative mechanisms; and (iv) altering cellular metabolism (Figure 1) [1,2].

 Functions of the endothelium. The endothelium is a dynamic monolayer of cells responsible for (a) vascular growth and formation [10-15], (b) haemostasis  and clot formation and degradation [16,17], (c) vital cellular processes [18], (d) providing a physical barrier to control permeability of molecules, peptides and cells  [19,20], (e) inflammatory response and leukocyte recruitment [21,22], and (f) haemodynamic balance through regulation of contraction and relaxation responses  [23,24].

Figure 1: Functions of the endothelium. The endothelium is a dynamic monolayer of cells responsible for (a) vascular growth and formation [10-15], (b) haemostasis and clot formation and degradation [16,17], (c) vital cellular processes [18], (d) providing a physical barrier to control permeability of molecules, peptides and cells [19,20], (e) inflammatory response and leukocyte recruitment [21,22], and (f) haemodynamic balance through regulation of contraction and relaxation responses [23,24].

Abbreviations: Acetyl-CoA: Acetyl Coenzyme A; ALM: Almandine; Ang: Angiopoietin; AngA: Angiotensin A; AngII: Angiotensin II; Ang(1-7): Angiotensin 1-7; AP-1: Activator Protein-1; ASNS: Asparagine Aspartate; AT: Antithrombin; ATP: Adenosine Triphosphate; AT1R: Angiotensin Type 1 Receptor; bFGF: basal Fibroblast Growth Factor; CD146: Cluster Differentiation 146; COX: Cyclooxygenase; DAMPs: Danger-Associated Molecular Patterns; DLL4: Delta-Like Ligand 4; EDHP: Endothelium-Derived Hyperpolarising Factor; eNOS: Endothelial Nitric Oxide Synthase; FGFR: Fibroblast Growth Factor Receptor; FAS: Fatty Acid Synthase; FGF: Fibroblast Growth Factor; GM-CSF: Granulocyte-Macrophage Colony-Stimulating Factor; GP: Glycerate 3-Phosphate; GPIb: Glycoprotein Ib; Hep: Heparin-like molecules; HGF: Hepatocyte Growth Factor; HIF: Hypoxia-Inducing Factor; HK: Hexokinase 2; HMGB1: High-Mobility Group Box-1; HSPG: Heparan Sulfate Proteoglycans; ICAM: Intracellular Adhesion Molecule; IGF: Insulin-Like Growth Factor; iNOS: inducible Nitric Oxide Synthase, IL-1: Interluikin-1, IL1R: Interluikin-1 Receptor; IRAK: IL-1R-Associated Kinase; LFA: Lymphocyte Function-Associated Antigen; MasR: MAS1 Oncogene Receptor; MCP-1: Monocyte Chemoattractant Protein-1; MLCK: Myosin-Light-Chain Kinase; MLP: Myosin Light Chain; MMPs: Matrix Metalloproteinases; MrgDR: MAS1 Oncogene, G-coupled Receptor; MyD88: Myeloid Differentiation Factor 88; NADH: Nicotinamide Adenine Dinucleotide Phosphate; NF-κB: Nuclear Factor Kappa B; NO: Nitric Oxide; Nox: NADPH Oxidase; PAF: Platelet-Activating Factor; PAI-1: Plasminogen Activator Inhibitor 1; PAMPs: Pattern-Associated Molecular Patterns; PAR: Protease Activating Receptor; PDGF: Platelet-Derived Growth Factor; PECAM: Platelet Endothelial Cell Adhesion Molecule; PGI2: Prostaglandin I2; PI3K: Phosphoinositide 3-Kinase; PLGF: Placental Growth Factor; PLG: Plasminogen; PLGR: Plasminogen Receptor; P2Y1: Purinergic Receptor P2Y G protein-coupled 1; RIP-1: Receptor-Interacting Protein 1; SAH: S-Adenosylhomocysteine; SAM: S-adenosylmethionine; TF: Tissue Factor; TFPI: Tissue Factor Pathway Inhibitor; TGF: Transforming Growth Factor; Tie: Angiopoietin Receptor; TIRAP: Toll/IL-1 Receptor Accessory Protein; TLR: Toll-Like Receptor; TM: Thrombomodulin; TNF: Tumor-Necrosis Factor; TNFR: TNF Receptor; TR: Thromboxane Receptor; TRADD: TNFR-Associated Via Death Domain Protein; TRAF: TNFR-Associated Factor; TXA2: Thromboxane; t-PA: tissue Plasminogen Activator; u-PA: urokinase Plasminogen Activator; VCAM: Vascular Cell Adhesion Molecule; VEGF: Vascular Endothelial Growth Factor; VEGFR: Vascular Endothelial Growth Factor Receptor; VRACs: Volume Regulated Anion Channels; vWF: von Willebrand Factor; Wnt: Wingless/integrated

Nitric oxide (NO) represents an important and potent endothelium-derived relaxing factor that is generated when the enzyme endothelial nitric oxide synthase (eNOS) catalases the conversion of L-arginine into L-citrulline and NO [1]. NO then diffuses into vascular smooth muscle cells where it activates guanylate cyclase, resulting in cyclic guanosine monophosphate (cGMP)-mediated vasodilation [1]. NO plays a central role in maintaining a quiescent state of the vascular wall by inhibiting mechanisms of inflammation, cell proliferation and thrombosis [1]. Additionally, the endothelium mediates hyperpolarization of vascular smooth muscle cells through NO-independent vasodilation pathway, resulting in augmented potassium conductance and depolarization of vascular smooth muscles, a compensatory mechanism that is favored during loss of NOmediated vasodilation [1]. A loss of endothelial barrier function, referred to as endothelial dysfunction, has been identified as the first vascular disturbance associated with underlying chronic inflammatory conditions, such as atherosclerosis, hyperhomocysteinemia and sepsis [3-5], and is defined as a systemic pathological state resulting from a plethora of cellular events, including diminished NO availability (partially due to decreased eNOS-derived NO synthesis), increased inflammation and oxidative stress, leukocyte infiltration and adhesion, and dysregulated endothelial cell metabolism [1,2].

This review focuses on mechanisms exerted by innate immunity and sensory nerves that drive endothelial dysfunction, the central pathophysiological process in life-threatening systemic inflammation. Specific emphasis is directed on the pathogenic features of extracellular high mobility group box protein 1 (HMGB1), a ubiquitous nuclear protein actively released as a proinflammatory mediator from numerous cell types, including sensory neurons during loss of tissue homeostasis [6-9]. Extracellular HMGB1 acts upstream of the classical proinflammatory cytokines and chemokines and all nucleated cells as well as platelets passively release HMGB1 extracellularly during lytic cell death.

Extracellular HMGB1 Biology

HMGB1 is a protein present in the nucleus of all animal cells and expresses 214 amino acid residues arranged in two positively charged DNA-binding domains and a negatively charged C-terminal region [6]. HMGB1 serves vital intranuclear functions required for transcription, nucleosome formation, DNA repair, and appropriate DNA conformation. Lytic cell death mediates passive extracellular HMGB1 release. Active extracellular HMGB1 release, induced by numerous exogenous and endogenous stimuli, occurs in multiple steps [25]. Two nuclear localization sites (NLS) secure a dominant nuclear HMGB1 localization during cellular homeostasis. Translocation of nuclear HMGB1 to the cytoplasm requires posttranslational molecular modifications. These changes include hyperacetylation of critical lysines in the two NLS sites, preventing the continuous bidirectional shuttle of HMGB1 between the cytoplasm and the nucleus and results in cytoplasmic HMGB1 accumulation [26]. Nuclear hyperacetylation is accomplished via increased histoneacetylase activity and decreased histone-deacetylase (HDAC) activity. Preventing hyperacetylation thus offers a therapeutic strategy to inhibit extracellular HMGB1 release. Cytoplasmic HMGB1 is released extracellularly via several mechanisms. One route proceeds via exocytosis of secretory lysosomes, a pathway also utilized for interleukin (IL)-1β secretion, although HMGB1 and IL-1β are stored in separate vesicles [27]. Another route for HMGB1 to exit cells takes place on the surface of microparticles derived from activated platelets [28]. Vascular injury induces massive extracellular release of HMGB1 from platelets displaying a central role in the pathogenesis of thrombosis formation and neutrophil activation [28]. Programmed, proinflammatory cell death (pyroptosis) is an additional mechanism for regulated HMGB1 release [29,30]. This process is a consequence of inflammasome activation and gasdermin D cleavage inducing pore formation, rupturing the outer cell membrane. This pathway is as an example of the dominant mechanism for HMGB1 release during gram-negative sepsis when caspase-1/caspase-11 doubledeficient mice express markedly reduced systemic HMGB1 levels [30-32]. Most importantly, recent research demonstrates that stimulated sensory neurons can actively secrete HMGB1 in an antidromic fashion by mechanisms not fully elucidated [7,8]. Nociceptors harvested from transgenic mice expressing channelrhodopsin-2 in neurons directly release HMGB1 by light exposure causing neuroinflammation in contrast to mice engineered with neuronal HMGB1 gene deficiency. A plausible functional role of HMGB1 released from sensory neurons in the vascular wall during systemic inflammation has not been studied so far but deserves future attention.

The redox state of HMGB1 determines the extracellular HMGB1 receptor usage [33]. The DNA-binding boxes in HMGB1 contain three cysteines and the redox state of each of these residues is key for the extracellular receptor association and activation [33]. All three cysteines reside in a fully reduced state with thiol groups (all-thiol HMGB1) and are present in quiescent cells. This isoform acts extracellularly as a chemotactic factor when operating together with C-X-C motif chemokine ligand (CXCL) 12 via C-X-C chemokine receptor (CXCR) 4 [34]. Mild HMGB1 oxidation generates disulfide HMGB1 characterized by a disulfide bond between Cys23 and Cys45 while keeping Cys106 in the reduced form. Disulfide HMGB1 is a potent proinflammatory molecule operating via toll-like receptor (TLR) 4 [35]. Like lipopolysaccharides (LPS), disulfide HMGB1 binds to myeloid differentiation factor-2 (MD-2) in the TLR4 receptor complex, but at a separate position. Further oxidation of HMGB1 produces sulfonyl groups on any or all cysteine residues creating sulfonyl HMGB1, which is a non-inflammatory isoform.

An additional and important proinflammatory HMGB1 receptor is the receptor for glycated end-products (RAGE) [36]. This is a multi-ligand receptor expressed by many cell types, including endothelial cells, predominantly as a preformed intracellular molecule available for prompt nuclear factor kappa B (NF-kB)-controlled translocation to the cell surface when cellular homeostasis is challenged. Extracellular HMGB1 readily forms heterocomplexes with multiple other extracellular damage-associated molecular pattern molecules (DAMPs) and pathogen-associated molecular pattern molecules (PAMPs) [37], which are endocytosed via RAGE for further transfer into the endolysosomal compartment [30,38] (Figure 2].

HMGB1 transports multiple extracellular DAMPs and PAMPs, here exemplified by LPS, into the inside of endothelial cells. HMGB1 has two LPS-binding sites  and thus forms an extracellular HMGB1–LPS complex that gets endocytosed via RAGE to the endolysosomal system. High intralysosomal levels of HMGB1 present at  low pH damages the lysosomal membrane enabling LPS to leak into the cytosol culminating in activation of the intracellular LPS sensor caspase-11 (caspases-4 and  ?5 in humans) that instigates inflammation and coagulation.

Figure 2: HMGB1 transports multiple extracellular DAMPs and PAMPs, here exemplified by LPS, into the inside of endothelial cells. HMGB1 has two LPS-binding sites and thus forms an extracellular HMGB1–LPS complex that gets endocytosed via RAGE to the endolysosomal system. High intralysosomal levels of HMGB1 present at low pH damages the lysosomal membrane enabling LPS to leak into the cytosol culminating in activation of the intracellular LPS sensor caspase-11 (caspases-4 and −5 in humans) that instigates inflammation and coagulation.

Abbreviations: HMGB1: High-Mobility Group Box-1; IL: Interleukin; LPS: Lipopolysaccharide; RAGE: Receptor For Advanced Glycated End-Products; TLR: Toll-Like Receptor.

Most of these DAMPs and PAMPs do not cause inflammation when present extracellularly since their cognate proinflammatory receptors are located intracellularly in target cells but not on cell surfaces. However, HMGB1 provides a transport intracellularly for these danger molecules via RAGE-mediated endocytosis and in addition by performing a unique function inside acidic lysosomes disrupting the lysosomal membrane at low pH allowing its transported molecules to circumvent degradation and leakage into the cytosol. Some of these imported molecules are potent ligands to inflammasomes, retinoic acid-inducible gene I, cGMPadenosine monophosphate synthase-stimulator of interferon genes, and additional receptors causing inflammation and coagulation.

RAGE expressed on endothelial cells encounters the pathogenic HMGB1 heterocomplexes during the early stage of systemic inflammation. A significant quantitative discrepancy between HMGB1 levels in arterial versus venous plasma samples has been reported in severe trauma patients [39]. Arterial HMGB1 concentration was consistently lower than venous concentration in simultaneously obtained samples (arterial = 0.6 x venous; 95% CI 0.30–0.90) [39]. Significant amounts of HMGB1 are thus hypothesized to be removed in pulmonary circulation, where there is constitutive luminal endothelial RAGE expression in contrast to other organs. The mechanism and reason for HMGB1 elimination remain to be clarified, but one may speculate that it represents a function to degrade PAMP and DAMP molecules carried by HMGB1 via RAGE mediated endocytosis to the lysosomes in endothelial cells in the pulmonary circulation. Many of these DAMPs and PAMPs share a poor ability to generate antibody responses. Physiological amounts of HMGB1 are insufficient to damage the lysosomal membrane allowing the expected and beneficial degradation of the HMGB1-partner molecule complexes, while pathologically increased intralysosomal HMGB1 levels cause lysosomal damage and leakage to the cytoplasm with subsequent inflammasome activation.

HMGB1 and Endothelial Dysfunction

Endothelial cells both release [40], and respond [41], to HMGB1 and results from in vivo and in vitro studies indicate that these biological activities have important pathophysiological consequences.

Extracellular HMGB1 Activates Endothelial Cells

Human microvascular endothelium cocultured with HMGB1 becomes activated in a dose- and time-dependent manner to increase the cell surface expression of intercellular adhesion molecule 1, vascular cell adhesion molecule 1, and HMGB1 receptors RAGE and TLR4 [41-45]. Furthermore, HMGB1 induces secretion of the proinflammatory cytokines and chemokines (e.g., tumor necrosis factor (TNF), IL-6, IL-8, and monocyte chemoattractant protein 1) and the critical regulators of fibrinolysis (e.g., plasminogen activator inhibitor 1 and tissue plasminogen activator). HMGB1 stimulation generates a transient phosphorylation of mitogen-activated protein kinases, extracellular signal-related kinase, Jun N-terminal kinase, and nuclear NF-kB translocation in the endothelial cells. Moreover, the pathogenic effect of extracellular HMGB1 is further highlighted by a recent study that reported HMGB1 as an important regulator of various endothelium-dependent activities in genetically modified mice that have an absence of endothelial HMGB1 [46]. Mice in this study displayed elevated mean arterial pressure, increased plasma HMGB1 concentration, reduced endothelium-dependent relaxation and decreased NO and eNOS phosphorylation and peptide expression [46].

Preclinical in vivo studies have demonstrated the ability of HMGB1 to induce human endothelial cell cytoskeletal rearrangement and barrier disruption via increasing paracellular gap formation in concert with loss of peripheral organized actin fibers, and dissociation of cell-cell junctional cadherins [47]. Thought-provoking clinical in vivo observations have been reported about a possible pathogenic role of HMGB1 in Kawasaki disease, a severe acute vasculitis in infancy and childhood. Kawasaki patients generally express high systemic HMGB1 levels and may develop endothelial damage mediating coronary endothelial dysfunction resulting in coronary artery aneurysms. Results from a recent clinical study indicated that nucleotidebinding domain, leucine-rich–containing family, pyrin domain– containing-3-dependent endothelial cell pyroptosis, activated by HMGB1/RAGE/cathepsin B signaling, represented a central mechanism in the pathophysiology [48]. Human umbilical vein endothelial cells cultured in plasma from Kawasaki patients developed pyroptosis, while the addition of neutralizing antiHMGB1 antibodies or RAGE-specific antagonists to the cell cultures prevented the pyroptosis and the subsequent release of IL-1a, IL-1b, IL-18, and HMGB1 [48].

Activated Endothelial Cells Release HMGB1

Like many other cell types, stressed endothelial cells release HMGB1 extracellularly via active mechanisms or via passive leakage occurring after lytic modes of cell death [49]. Sterile inflammation and infections are potent stimuli for the generation of active HMGB1 release from endothelial cells. Chemical modifications including hyperacetylation and phosphorylation of HMGB1 in the nucleus are necessary initial steps to allow nuclear HMGB1 export to the cytoplasm for further extracellular secretion [25]. Antagonizing the nuclear modifications of HMGB1 offers a therapeutic anti-inflammatory strategy that has been successfully used in preclinical models of severe systemic inflammatory disorders, but not yet in patients. Activating the endogenous cholinergic anti-inflammatory pathway is one such therapeutic example that will be outlined later in this review. Administration of histidine-rich glycoprotein (HRG), a 75 kDa plasma protein, has been demonstrated to improve the survival rate of septic mice through the regulation of neutrophils and endothelium barrier function [42]. The role of HRG in endothelial barrier protection was concluded to depend mainly on its effect of inhibiting HMGB1 release from endothelial cells.

HMGB1 Causes Blood Brain Barrier Disruption

The blood-brain barrier (BBB) is composed of vascular endothelial cells with tight junctions, pericytes, extracellular matrix, and endfeet of astrocytic processes [42,44,50-52]. The critical function of the BBB is to protect the central nervous system from the entry of molecules that might harm the brain. High levels of systemic as well as extracellular cerebral HMGB1 have been demonstrated to substantially increase BBB permeability, in association with morphological contractile changes in vascular endothelial cells and pericytes [42,44,50- 52]. Nishibori and colleagues have demonstrated that systemic administration of anti-HMGB1 monoclonal antibodies (mAb) protected the BBB integrity and inhibited the translocation of HMGB1 from neurons in experimental models of cerebral ischemia or hemorrhage [53-55]. It seems conceivable that the initial event for the protection of the BBB integrity occurred via antibody mediated HMGB1 neutralization close to the capillary endothelial cells. Using a rat-reconstituted BBB in vitro system composed of vascular endothelial cells, pericytes and astrocytes, the same research group confirmed that recombinant HMGB1 incubation caused a contractile response in endothelial cells and pericytes, which was inhibited by anti-HMGB1 mAb [53].

Does HMGB1 Released From Vascular Sensory Nerves Cause Endothelial Dysfunction?

The significance of the role of vascular nerves in the development of endothelial dysfunction in systemic inflammatory diseases remains poorly understood. Vasomotor control is exerted by sympathetic vasoconstrictor nerves in combination with vasodilator influences from transient receptor potential vanilloid subfamily member 1 (TRPV1)-expressing sensory-motor nerve release of substance P and calcitonin generelated peptide , both of which are neurotransmitters that also possess proinflammatory effects [56]. These nerves run close to each other in the vascular wall and are located between the tunica media and the tunica adventitia, but they do not directly innervate the endothelial cells. However, there is strong evidence from different types of studies that microvascular smooth muscle is electrically coupled to the endothelium and that smooth muscle activation functionally affects endothelial cells [57-60]. This is particularly the case in the microcirculation, where the distance between the media-adventitia border and the tunica intima is minor. Recent discoveries demonstrate that TRPV1-positive sensory neurons also actively release HMGB1 in a retrograde manner and upregulate inflammation in the periphery [7,8]. Optogenetic stimulation of engineered peripheral sensory nerves induces active HMGB1 release and inflammation, while neuronal HMGB1 knock-out or HMGB1 neutralization via antibodies inhibit inflammation in models of arthritis and nerve injury. Analogous studies have not yet been performed in vascular pathology but should be conducted since the results may have important clinical therapeutic implications.

Targeting HMGB1 to Ameliorate Endothelial Dysfunction

Recognition of HMGB1 as a critical molecule in the pathogenesis of dysfunctional microcirculation in systemic inflammatory conditions makes it a valid therapeutic target. There are several successful approaches that have been studied in preclinical models and they will be further discussed. 

Activating the Endogenous Cholinergic Anti-Inflammatory Pathway to Inhibit HMGB1 Release and to Reduce RAGEMediated Endocytosis of HMGB1-DAMP/PAMP Complexes: Inflammation is regulated by the nervous system, but this insight has so far not led to any clinical therapy for sepsis. Neuronally released HMGB1 accelerates inflammation, while acetylcholine, released via the cholinergic anti-inflammatory pathway, in contrast inhibits excessive inflammation [61,62]. Acetylcholine is released in all vagally innervated, organs including the celiac ganglion, where it activates the catecholaminergic splenic nerve to discharge norepinephrine in the spleen to stimulate a subset of splenic T lymphocytes capable of acetylcholine synthesis [63]. These activated T cells may then leave the spleen and function like mobile neurons to release acetylcholine that will down-regulate inflammation in body compartments that lack vagal innervation. The cholinergic mechanism inhibits proinflammatory HMGB1 release via signals that require acetylcholine and alpha-7 nicotinic acetylcholine receptors (α7nAchR) (Figure 3A) [64, 65].

HMGB1 antagonists ameliorate HMGB1-dependent inflammation via separate mechanisms.

Figure 3 :HMGB1 antagonists ameliorate HMGB1-dependent inflammation via separate mechanisms.

Abbreviations: α7nAchR: Alpha-7 Nicotinic Acetylcholine Receptors; DAMP: Danger-Associated Molecular Pattern; HMGB1: High-Mobility Group Box-1; PAMP: Pattern-Associated Molecular Pattern; RAGE: Receptor For Advanced Glycated End-Products; SIRT1: Sirutin 1

Acetylation of multiple lysines in HMGB1 is necessary for the nuclear HMGB1 export needed for further active extracellular HMGB1 release [26]. Histone deacetylases, including sirutin 1 (SIRT1), prevent nuclear HMGB1 export. Acetylcholine boosts SIRT1 activity via α7nAChR and promotes nuclear HMGB1 sequestration [66]. HMGB1 is also dependent on NF-kB assistance for the cytoplasmic translocation and SIRT1 inhibits NF-κB activity by deacetylating the RelA/p65 subunit [67]. In addition, acetylcholine inhibits RAGE-mediated endocytosis of HMGB1 complexes, and TLR4/myeloid differentiation factor 88 (MyD88]/ NF-κB signaling via a7nAChR interaction [68,69]. Electrical vagus nerve stimulation (VNS) as well as systemic administration of a7nAChR-specific agonists has been demonstrated to improve the outcome in preclinical endotoxemia and sepsis models [64,70-74]. A need for surgical VNS device implantation can be circumvented by transcutaneous auricular VNS (taVNS) using an external pulse generator [75,76]. The auricular branch of the vagus nerve reaches superficial parts in the concha and tragus in both ears, enabling taVNS as a safer therapeutic alternative to an implanted VNS device. This approach has been successfully tested in murine polymicrobial sepsis resulting in reduced lethality and systemic HMGB1 levels [77].

Activation of α7nAChR can also be achieved via systemic administration of α7nAChR-specific agonists. One such molecule termed GTS-21 dose-dependently suppressed NF-kB activation and inhibited TNF and HMGB1 release from endotoxin-activated mouse macrophages [64]. In vivo, GTS-21 significantly reduced serum HMGB1 levels and improved the survival rate in mice with gram-negative polymicrobial severe sepsis. The compound has been therapeutically administered safely and well tolerated to patients with schizophrenia in pilot studies [78,79]. Further studies are warranted to translate the beneficial effects via the cholinergic anti-inflammatory pathway into clinical use for patients with severe systemic inflammation and endothelial dysfunction.

Administration of Low-Dose Heparin or Non Anticoagulant Heparin: Heparin is a negatively charged polysaccharide that binds to specific, positively charged sites in HMGB1 and prevents HMGB1 from binding to RAGE and thus restricts HMGB1-mediated import in endothelial cells of inflammasome-activating molecules (Figure 3B) [80,81]. Furthermore, non-anticoagulant heparin also binds HMGB1 as well as extracellular histones and prevents histone-mediated cytotoxicity, another dominant pathogenic route in severe sepsis. The effective doses of heparin required to inhibit LPS–HMGB1 toxicity and improve survival in experimental Gram-negative sepsis are much lower than those needed for the anticoagulant effects [80].

The endothelial glycocalyx is a gel-like layer composed of a network of proteoglycans, glycoproteins, and glycolipids at the luminal side of endothelial cells lining the blood vessels throughout the body. Proinflammatory molecules including HMGB1 may disrupt the endothelial glycocalyx, leading to edema, increased macromolecular leakage, and cytosolic LPS delivery. It is thus of therapeutic interest that heparin as well as nonanticoagulant heparin prevent the degradation of glycocalyx [80]. Modified heparin without anticoagulant activity also prevents LPS from binding to HMGB1 and prevents RAGE-mediated endocytosis of HMGB1 and of HMGB1 heterocomplexes. One study reporting successful heparin treatment in experimental Gramnegative sepsis concurrently found that the identical strategy failed in Staphylococcus aureus-induced sepsis, indicating that the pathogenic mechanisms operating in Gram-positive versus Gram-negative sepsis are different [80]. Staphylococcal and Streptococcal superantigens induce separate cytokine patterns than LPS with possibly diverse effects on endothelial functions [82,83]. Future clinical studies of heparin treatment in sepsis thus preferably need stratification of patients based on an identification of invading pathogens. There are several clinical studies on the therapeutic efficacy of heparin in treating sepsis with conflicting results, possibly due to the lack of stratification.

Systemic Treatment with Soluble Thrombomodulin: Thrombomodulin is an endothelial cell surface thrombin receptor that converts thrombin into an anticoagulant. During the hypercoagulable state after endothelial injury, thrombomodulin is released into the intravascular space by proteolytic cleavage of the endothelium component [84]. Soluble thrombomodulin binds and neutralizes extracellular histones and HMGB1 and supports the proteolytic thrombin-mediated degradation of HMGB1 (Figure 3C) [85]. Recombinant soluble thrombomodulin has been shown to protect patients with disseminated intravascular coagulation after severe trauma or in severe sepsis [86,87].

DISCUSSION

Based on the presented preclinical results, HMGB1 is a valid and logical therapeutic target molecule to study to further ameliorate the consequences of endothelial dysfunction in severe systemic inflammatory diseases. However, this mission signifies a distinct clinical challenge for several reasons. Extracellular HMGB1 causes endothelial dysfunction via several separate pathways operating in concert. These paths include CXCR4, TLR4, and RAGE and they may possibly need to be counteracted by distinct therapeutic approaches. The general understanding of the relative functional importance of each of these receptormediated activities in any given severe systemic inflammatory clinical disease is presently insufficient. Another problem, based on steric hindrance for HMGB1-binding antagonists, is provoked by the exceptional readiness of extracellular HMGB1 to form heterocomplexes in vivo with other molecules.

An alternative therapeutic approach, that partially circumvents the discussed limitations, is to reduce the released amounts of HMGB1, occurring passively after lytic cell death or after active cellular secretion. The half-life time of plasma HMGB1 passively released after necrosis in severe trauma patients is 26 min and this early, transient peak of extracellular all-thiol HMGB1 has not been ascertained to generate subsequent severe systemic inflammatory clinical problems [88]. However, trauma patients who go on to develop systemic inflammatory response syndrome (SIRS), produce a second extracellular HMGB1-wave peaking 3-6 hours after the initiating trauma [88]. The cellular source of this secondary HMGB1 release has not been identified but is most likely the result of active HMGB1 secretion. This is a typical example where therapeutic intervention with agents preventing active extracellular HMGB1 release should be important to study. Inhibiting translocation of the nuclear pool of HMGB1 to the cytoplasm in stressed cells is conceivably one way to accomplish this. Promoting histone deacetylase activities inhibit nuclear HMGB1 hyperacetylation and thus prevents HMGB1 from entering the cytoplasm for further extracellular release. Agents that enhance SIRT1- and other HDAC-activities include acetylcholine [66,89,90], metformin [91], resveratrol [92], statins [93], and additional compounds. Strategies aimed to inhibit specific post-translational modifications of HMGB1 needed for nuclear HMGB1 export may conceivably provide novel opportunities to ameliorate endothelial dysfunction in patients with severe inflammation.

AUTHOR CONTRIBUTIONS

Conceptualization, U.A.; writing original draft preparation, H.Y., L.K.G., A.Z. and U.A.; and writing review and editing, H.Y. and U.A. All authors have read and agreed to this submitted version of the manuscript.

FUNDING

Susan Claster educational advancement award from Advancing Women in Science and Medicine (AWSM, The Feinstein Institutes for Medical Research, Northwell Health, NY, to HY).

ACKNOWLEDGMENTS

L.K.G. and A.Z. would like to acknowledge the Institute for Health and Sport, Victoria University, for their support as L.K.G. is a recipient of Victoria University postgraduate scholarships.

REFERENCES

1. Deanfield JE, Halcox JP, Rabelink TJ. Endothelial function and dysfunction: testing and clinical relevance. Circulation. 2007; 115: 1285-95.

2. Xu S, Ilyas I, Little PJ, Li H, Kamato D, Zheng X, et al. Endothelial dysfunction in atherosclerotic cardiovascular diseases and beyond: from mechanism to pharmacotherapies. Pharmacol Rev. 2021; 73: 924-67.

3. Joffre J, Hellman J, Ince C, Ait-Oufella H. Endothelial Responses in Sepsis. Am J Respir Crit Care Med. 2020; 202: 361-70.

4. Ince C, Mayeux PR, Nguyen T, Gomez H, Kellum JA, Ospina-Tascón GA, et al. THE ENDOTHELIUM IN SEPSIS. Shock. 2016; 45: 259-70.

5. Opal SM, van der Poll T. Endothelial barrier dysfunction in septic shock. J Intern Med. 2015; 277: 277-93.

6. Kang R, Chen R, Zhang Q, Hou W, Wu S, Cao L, et al. HMGB1 in health and disease. Mol Aspects Med. 2014; 40: 1-116.

7. Yang H, Zeng Q, Silverman HA, Gunasekaran M, George SJ, Devarajan A, et al. HMGB1 released from nociceptors mediates inflammation. Proc Natl Acad Sci U S A. 2021; 118: e2102034118.

8. Yang H, Andersson U, Brines M. Neurons Are a Primary Driver of Inflammation via Release of HMGB1. Cells. 2021; 10: 2791.

9. Andersson U. Hyperinflammation: On the pathogenesis and treatment of macrophage activation syndrome. Acta paediatr. 2021; 110: 2717-22.

10. Carmeliet P, Jain RK. Molecular mechanisms and clinical applications of angiogenesis. Nature. 2011; 473: 298-307.

11. Carmeliet P. Mechanisms of angiogenesis and arteriogenesis. Nat Med. 2000; 6: 389-95.

12. Jourde-Chiche N, Fakhouri F, Dou L, Bellien J, Burtey S, Frimat M, et al. Endothelium structure and function in kidney health and disease. Nat Rev Nephrol. 2019; 15: 87-108.

13. Heil M, Schaper W. Influence of mechanical, cellular, and molecular factors on collateral artery growth (arteriogenesis). Circ Res. 2004; 95: 449-58.

14. Korhonen EA, Murtomäki A, Jha SK, Anisimov A, Pink A, Zhang Y, et al. Lymphangiogenesis requires Ang2/Tie/PI3K signaling for VEGFR3 cell-surface expression. J Clin Invest. 2022; 132: e155478.

15. Deng Y, Zhang X, Simons M. Molecular controls of lymphatic VEGFR3 signaling. Arterioscler Thromb Vasc Biol. 2015; 35: 421-9.

16. Neubauer K, Zieger B. Endothelial cells and coagulation. Cell Tissue Res. 2022; 387: 391-8.

17. Chapin JC, Hajjar KA. Fibrinolysis and the control of blood coagulation. Blood Rev. 2015; 29: 17-24.

18. Falkenberg KD, Rohlenova K, Luo Y, Carmeliet P. The metabolic engine of endothelial cells. Nat Metab. 2019; 1: 937-46.

19. Claesson-Welsh L. Vascular permeability—the essentials. Ups J Med Sci. 2015; 120: 135-43.

20. Cai A, Chatziantoniou C, Calmont A. Vascular permeability: regulation pathways and role in kidney diseases. Nephron. 2021; 145: 297-310.

21. Pober JS, Sessa WC. Evolving functions of endothelial cells in inflammation. Nat Rev Immunol. 2007; 7: 803-15.

22. Rao RM, Yang L, Garcia-Cardena G, Luscinskas FW. Endothelialdependent mechanisms of leukocyte recruitment to the vascular wall. Circ Res. 2007; 101: 234-47.

23. Gomez-Alamillo C, Juncos LA, Cases A, Haas JA, Romero JC. Interactions between vasoconstrictors and vasodilators in regulating hemodynamics of distinct vascular beds. Hypertension. 2003; 42: 831-6.

24. Paz Ocaranza M, Riquelme JA, García L, Jalil JE, Chiong M, Santos RA, et al. Counter-regulatory renin–angiotensin system in cardiovascular disease. Nat Rev Cardiol. 2020; 17: 116-29.

25. Kwak MS, Kim HS, Lee B, Kim YH, Son M, Shin JS. Immunological Significance of HMGB1 Post-Translational Modification and Redox Biology. Front Immunol. 2020; 11: 1189.

26. Bonaldi T, Talamo F, Scaffidi P, Ferrera D, Porto A, Bachi A, et al. Monocytic cells hyperacetylate chromatin protein HMGB1 to redirect it towards secretion. EMBO J. 2003; 22: 5551-60. 

27. Gardella S, Andrei C, Ferrera D, Lotti LV, Torrisi MR, Bianchi ME, et al. The nuclear protein HMGB1 is secreted by monocytes via a nonclassical, vesicle-mediated secretory pathway. EMBO Rep. 2002; 3: 995-1001.

28. Bernard NJ. HMGB1(+) platelet microparticles damage the endothelium. Nat Rev Rheumatol. 2018; 14: 499.

29. Lamkanfi M, Sarkar A, Vande Walle L, Vitari AC, Amer AO, Wewers MD, et al. Inflammasome-dependent release of the alarmin HMGB1 in endotoxemia. J Immunol. 2010; 185: 4385-92.

30. Deng M, Tang Y, Li W, Wang X, Zhang R, Zhang X, et al. The Endotoxin Delivery Protein HMGB1 Mediates Caspase-11-Dependent Lethality in Sepsis. Immunity. 2018; 49: 740-53.e7.

31. Hagar JA, Powell DA, Aachoui Y, Ernst RK, Miao EA. Cytoplasmic LPS activates caspase-11: implications in TLR4-independent endotoxic shock. Science. 2013; 341: 1250-3.

32. Kayagaki N, Warming S, Lamkanfi M, Vande Walle L, Louie S, Dong J, et al. Non-canonical inflammasome activation targets caspase-11. Nature. 2011; 479: 117-21.

33. Yang H, Lundbäck P, Ottosson L, Erlandsson-Harris H, Venereau E, Bianchi ME, et al. Redox modifications of cysteine residues regulate the cytokine activity of HMGB1. Mol Med. 2021; 27: 58.

34. Schiraldi M, Raucci A, Muñoz LM, Livoti E, Celona B, Venereau E, et al. HMGB1 promotes recruitment of inflammatory cells to damaged tissues by forming a complex with CXCL12 and signaling via CXCR4. J Exp Med. 2012; 209: 551-63.

35. Yang H, Wang H, Ju Z, Ragab AA, Lundback P, Long W, et al. MD-2 is required for disulfide HMGB1-dependent TLR4 signaling. J Exp Med. 2015; 212: 5-14.

36. Rauvala H, Rouhiainen A. RAGE as a receptor of HMGB1 (Amphoterin): roles in health and disease. Curr Mol Med. 2007; 7: 725-34.

37. Bianchi ME. HMGB1 loves company. J Leukoc Biol. 2009; 86: 573-6.

38. Xu J, Jiang Y, Wang J, Shi X, Liu Q, Liu Z, et al. Macrophage endocytosis of high-mobility group box 1 triggers pyroptosis. Cell Death Differ. 2014; 21: 1229-39.

39. Ottestad W, Rognes IN, Skaga E, Frisvoll C, Haraldsen G, Eken T, et al. HMGB1 concentration measurements in trauma patients: assessment of pre-analytical conditions and sample material. Mol Med. 2019; 26: 5.

40. Mullins GE, Sunden-Cullberg J, Johansson AS, Rouhiainen A, Erlandsson-Harris H, Yang H, et al. Activation of human umbilical vein endothelial cells leads to relocation and release of high-mobility group box chromosomal protein 1. Scand J Immunol. 2004; 60: 566- 73.

41. Fiuza C, Bustin M, Talwar S, Tropea M, Gerstenberger E, Shelhamer JH, et al. Inflammation-promoting activity of HMGB1 on human microvascular endothelial cells. Blood. 2003; 101: 2652-60.

42. Gao S, Wake H, Sakaguchi M, Wang D, Takahashi Y, Teshigawara K, et al. Histidine-Rich Glycoprotein Inhibits High-Mobility Group Box-1- Mediated Pathways in Vascular Endothelial Cells through CLEC-1A. iScience. 2020; 23: 101180.

43. Nishibori M, Mori S, Takahashi HK. Anti-HMGB1 monoclonal antibody therapy for a wide range of CNS and PNS diseases. J Pharmacol Sci. 2019; 140: 94-101.

44. Nishibori M, Wang D, Ousaka D, Wake H. High Mobility Group Box-1 and Blood-Brain Barrier Disruption. Cells. 2020; 9: 2650.

45. Namba T, Tsuge M, Yashiro M, Saito Y, Liu K, Nishibori M, et al. Anti-high mobility group box 1 monoclonal antibody suppressed hyper-permeability and cytokine production in human pulmonary endothelial cells infected with influenza A virus. Inflamm Res. 2021; 70: 1101-11.

46. Zhou Q, Tu T, Tai S, Tang L, Yang H, Zhu Z. Endothelial specific deletion of HMGB1 increases blood pressure and retards ischemia recovery through eNOS and ROS pathway in mice. Redox Biol. 2021; 41: 101890.

47. Wolfson RK, Chiang ET, Garcia JG. HMGB1 induces human lung endothelial cell cytoskeletal rearrangement and barrier disruption. Microvasc Res. 2011; 81: 189-97.

48. Jia C, Zhang J, Chen H, Zhuge Y, Chen H, Qian F, et al. Endothelial cell pyroptosis plays an important role in Kawasaki disease via HMGB1/ RAGE/cathespin B signaling pathway and NLRP3 inflammasome activation. Cell Death Dis. 2019; 10: 778.

49. Chen R, Kang R, Tang D. The mechanism of HMGB1 secretion and release. Exp Mol Med. 2022; 54: 91-102.

50. Festoff BW, Sajja RK, van Dreden P, Cucullo L. HMGB1 and thrombin mediate the blood-brain barrier dysfunction acting as biomarkers of neuroinflammation and progression to neurodegeneration in Alzheimer’s disease. J Neuroinflammation. 2016; 13: 194.

51. Liang Y, Song P, Chen W, Xie X, Luo R, Su J, et al. Inhibition of Caspase-1 Ameliorates Ischemia-Associated Blood-Brain Barrier Dysfunction and Integrity by Suppressing Pyroptosis Activation. Front Cell Neurosci. 2020; 14: 540669.

52. Liu L, Wang N, Kalionis B, Xia S, He Q. HMGB1 plays an important role in pyroptosis induced blood brain barrier breakdown in diabetesassociated cognitive decline. J Neuroimmunol. 2022; 362: 577763.

53. Zhang J, Takahashi HK, Liu K, Wake H, Liu R, Maruo T, et al. Anti-high mobility group box-1 monoclonal antibody protects the blood-brain barrier from ischemia-induced disruption in rats. Stroke. 2011; 42: 1420-8.

54. Liu K, Mori S, Takahashi HK, Tomono Y, Wake H, Kanke T, et al. Antihigh mobility group box 1 monoclonal antibody ameliorates brain infarction induced by transient ischemia in rats. FASEB J. 2007; 21: 3904-16.

55. Wang D, Liu K, Wake H, Teshigawara K, Mori S, Nishibori M. Antihigh mobility group box-1 (HMGB1) antibody inhibits hemorrhageinduced brain injury and improved neurological deficits in rats. Sci Rep. 2017; 7: 46243.

56. Aalkjær C, Nilsson H, De Mey JGR. Sympathetic and Sensory-Motor Nerves in Peripheral Small Arteries. Physiol Rev. 2021; 101: 495-544.

57. Dora KA, Doyle MP, Duling BR. Elevation of intracellular calcium in smooth muscle causes endothelial cell generation of NO in arterioles. Proc Natl Acad Sci U S A. 1997; 94: 6529-34.

58. Jackson WF, Boerman EM, Lange EJ, Lundback SS, Cohen KD. Smooth muscle alpha1D-adrenoceptors mediate phenylephrine-induced vasoconstriction and increases in endothelial cell Ca2+ in hamster cremaster arterioles. Br J Pharmacol. 2008; 155: 514-24.

59. Kerr PM, Tam R, Ondrusova K, Mittal R, Narang D, Tran CH, et al. Endothelial feedback and the myoendothelial projection. Microcirculation. 2012; 19: 416-22.

60. Rahman A, Hughes A, Matchkov V, Nilsson H, Aalkjaer C. Antiphase oscillations of endothelium and smooth muscle [Ca2+]i in vasomotion of rat mesenteric small arteries. Cell calcium. 2007; 42: 536-47.

61. Alen NV. The cholinergic anti-inflammatory pathway in humans: State-of-the-art review and future directions. Neurosci Biobehav Rev. 2022; 136: 104622.

62. Tracey KJ. Physiology and immunology of the cholinergic antiinflammatory pathway. J Clin Invest. 2007; 117: 289-96.

63. Rosas-Ballina M, Olofsson PS, Ochani M, Valdés-Ferrer SI, Levine YA, Reardon C, et al. Acetylcholine-synthesizing T cells relay neural signals in a vagus nerve circuit. Science. 2011; 334: 98-101.

64. Pavlov VA, Ochani M, Yang LH, Gallowitsch-Puerta M, Ochani K, Lin X, et al. Selective alpha7-nicotinic acetylcholine receptor agonist GTS21 improves survival in murine endotoxemia and severe sepsis. Crit Care Med. 2007; 35: 1139-44.

65. Wazea SA, Wadie W, Bahgat AK, El-Abhar HS. Galantamine anti-colitic effect: Role of alpha-7 nicotinic acetylcholine receptor in modulating Jak/STAT3, NF-κB/HMGB1/RAGE and p-AKT/Bcl-2 pathways. Sci Rep. 2018; 8: 5110.

66. Li DJ, Huang F, Ni M, Fu H, Zhang LS, Shen FM. α7 Nicotinic Acetylcholine Receptor Relieves Angiotensin II-Induced Senescence in Vascular Smooth Muscle Cells by Raising Nicotinamide Adenine Dinucleotide-Dependent SIRT1 Activity. Arterioscler Thromb Vasc Biol. 2016; 36: 1566-76.

67. Sun HJ, Xiong SP, Cao X, Cao L, Zhu MY, Wu ZY, et al. Polysulfidemediated sulfhydration of SIRT1 prevents diabetic nephropathy by suppressing phosphorylation and acetylation of p65 NF-κB and STAT3. Redox Biol. 2021; 38: 101813.

68. Yang H, Liu H, Zeng Q, Imperato GH, Addorisio ME, Li J, et al. Inhibition of HMGB1/RAGE-mediated endocytosis by HMGB1 antagonist box A, anti-HMGB1 antibodies, and cholinergic agonists suppresses inflammation. Mol Med. 2019; 25: 13.

69. Zi SF, Li JH, Liu L, Deng C, Ao X, Chen DD, et al. Dexmedetomidinemediated protection against septic liver injury depends on TLR4/ MyD88/NF-kappaB signaling downregulation partly via cholinergic anti-inflammatory mechanisms. Int Immunopharmacolcology. 2019; 76: 105898.

70. Wang H, Liao H, Ochani M, Justiniani M, Lin X, Yang L, et al. Cholinergic agonists inhibit HMGB1 release and improve survival in experimental sepsis. Nat Med. 2004; 10: 1216-21.

71. Johnson RL, Wilson CG. A review of vagus nerve stimulation as a therapeutic intervention. J Inflamm Res. 2018; 11: 203-13.

72. Mihaylova S, Killian A, Mayer K, Pullamsetti SS, Schermuly R, Rosengarten B. Effects of anti-inflammatory vagus nerve stimulation on the cerebral microcirculation in endotoxinemic rats. J Neuroinflammation. 2012; 9: 183.

73. van Westerloo DJ, Giebelen IA, Meijers JC, Daalhuisen J, de Vos AF, Levi M, et al. Vagus nerve stimulation inhibits activation of coagulation and fibrinolysis during endotoxemia in rats. J Thromb Haemost. 2006; 4: 1997-2002.

74. Andersson U. The cholinergic anti-inflammatory pathway alleviates acute lung injury. Mol Med. 2020; 26: 64.

75. Pavlov VA, Chavan SS, Tracey KJ. Molecular and Functional Neuroscience in Immunity. Annu Rev Immunol. 2018; 36: 783-812.

76. Aranow C, Atish-Fregoso Y, Lesser M, Mackay M, Anderson E, Chavan S, et al. Transcutaneous auricular vagus nerve stimulation reduces pain and fatigue in patients with systemic lupus erythematosus: a randomised, double-blind, sham-controlled pilot trial. Ann Rheum Dis. 2021; 80: 203-8.

77. Huston JM, Gallowitsch-Puerta M, Ochani M, Ochani K, Yuan R, RosasBallina M, et al. Transcutaneous vagus nerve stimulation reduces serum high mobility group box 1 levels and improves survival in murine sepsis. Crit Care Med. 2007; 35: 2762-8.

78. Freedman R, Olincy A, Buchanan RW, Harris JG, Gold JM, Johnson L, et al. Initial phase 2 trial of a nicotinic agonist in schizophrenia. Am J Psychiatry. 2008; 165: 1040-7.

79. Olincy A, Harris JG, Johnson LL, Pender V, Kongs S, Allensworth D, et al. Proof-of-concept trial of an alpha7 nicotinic agonist in schizophrenia. Arch Gen Psychiatry. 2006; 63: 630-8.

80. Tang Y, Wang X, Li Z, He Z, Yang X, Cheng X, et al. Heparin prevents caspase-11-dependent septic lethality independent of anticoagulant properties. Immunity. 2021; 54: 454-67.e6.

81. Li L, Ling Y, Huang M, Yin T, Gou SM, Zhan NY, et al. Heparin inhibits the inflammatory response induced by LPS and HMGB1 by blocking the binding of HMGB1 to the surface of macrophages. Cytokine. 2015; 72: 36-42.

82. Andersson J, Nagy S, Björk L, Abrams J, Holm S, Andersson U. Bacterial toxin-induced cytokine production studied at the single-cell level. Immunol Rev. 1992; 127: 69-96.

83. Björk L, Andersson J, Ceska M, Andersson U. Endotoxin and Staphylococcus aureus enterotoxin A induce different patterns of cytokines. Cytokine. 1992; 4: 513-9.

84. Watanabe-Kusunoki K, Nakazawa D, Ishizu A, Atsumi T. Thrombomodulin as a Physiological Modulator of Intravascular Injury. Front Immunol. 2020; 11: 575890.

85. Abeyama K, Stern DM, Ito Y, Kawahara K, Yoshimoto Y, Tanaka M, et al. The N-terminal domain of thrombomodulin sequesters highmobility group-B1 protein, a novel antiinflammatory mechanism. J Clin Invest. 2005; 115: 1267-74.

86. Ito T, Kakihana Y, Maruyama I. Thrombomodulin as an intravascular safeguard against inflammatory and thrombotic diseases. Expert Opin Ther Targets. 2016; 20: 151-8.

87. Yamakawa K, Murao S, Aihara M. Recombinant Human Soluble Thrombomodulin in Sepsis-Induced Coagulopathy: An Updated Systematic Review and Meta-Analysis. Thromb Haemost. 2019; 119: 56-65.

88. Ottestad W, Rognes IN, Pischke SE, Mollnes TE, Andersson U, Eken T. Biphasic Release of the Alarmin High Mobility Group Box 1 Protein Early After Trauma Predicts Poor Clinical Outcome. Crit Care Med. 2019; 47: e614-e22.

89. Hwang JS, Choi HS, Ham SA, Yoo T, Lee WJ, Paek KS, et al. Deacetylationmediated interaction of SIRT1-HMGB1 improves survival in a mouse model of endotoxemia. Sci Rep. 2015; 5: 15971.

90. Wei S, Gao Y, Dai X, Fu W, Cai S, Fang H, et al. SIRT1-mediated HMGB1 deacetylation suppresses sepsis-associated acute kidney injury. Am J Physiol Renal Physiol. 2019; 316: F20-f31.

91. Cuyàs E, Verdura S, Llorach-Parés L, Fernández-Arroyo S, Joven J, Martin-Castillo B, et al. Metformin Is a Direct SIRT1-Activating Compound: Computational Modeling and Experimental Validation. Front Endocrinol (Lausanne) 2018; 9: 657.

92. Yu S, Zhou X, Xiang H, Wang S, Cui Z, Zhou J. Resveratrol Reduced Liver Damage After Liver Resection in a Rat Model by Upregulating Sirtuin 1 (SIRT1) and Inhibiting the Acetylation of High Mobility Group Box 1 (HMGB1). Med Sci Monit. 2019; 25: 3212-20.

93. Khayatan D, Razavi SM, Arab ZN, Khanahmadi M, Momtaz S, Butler AE, et al. Regulatory Effects of Statins on SIRT1 and Other Sirtuins in Cardiovascular Diseases. Life (Basel). 2022; 12: 760.

Yang H, Gadanec LK, Zulli A, Andersson U. (2023) HMGB1 as a Therapeutic Target to Alleviate Endothelial Dysfunction in Severe Systemic Inflammation. Ann Vasc Med Res 10(3): 1164.

Received : 06 Jul 2023
Accepted : 31 Aug 2023
Published : 31 Aug 2023
Journals
Annals of Otolaryngology and Rhinology
ISSN : 2379-948X
Launched : 2014
JSM Schizophrenia
Launched : 2016
Journal of Nausea
Launched : 2020
JSM Internal Medicine
Launched : 2016
JSM Hepatitis
Launched : 2016
JSM Oro Facial Surgeries
ISSN : 2578-3211
Launched : 2016
Journal of Human Nutrition and Food Science
ISSN : 2333-6706
Launched : 2013
JSM Regenerative Medicine and Bioengineering
ISSN : 2379-0490
Launched : 2013
JSM Spine
ISSN : 2578-3181
Launched : 2016
Archives of Palliative Care
ISSN : 2573-1165
Launched : 2016
JSM Nutritional Disorders
ISSN : 2578-3203
Launched : 2017
Annals of Neurodegenerative Disorders
ISSN : 2476-2032
Launched : 2016
Journal of Fever
ISSN : 2641-7782
Launched : 2017
JSM Bone Marrow Research
ISSN : 2578-3351
Launched : 2016
JSM Mathematics and Statistics
ISSN : 2578-3173
Launched : 2014
Journal of Autoimmunity and Research
ISSN : 2573-1173
Launched : 2014
JSM Arthritis
ISSN : 2475-9155
Launched : 2016
JSM Head and Neck Cancer-Cases and Reviews
ISSN : 2573-1610
Launched : 2016
JSM General Surgery Cases and Images
ISSN : 2573-1564
Launched : 2016
JSM Anatomy and Physiology
ISSN : 2573-1262
Launched : 2016
JSM Dental Surgery
ISSN : 2573-1548
Launched : 2016
Annals of Emergency Surgery
ISSN : 2573-1017
Launched : 2016
Annals of Mens Health and Wellness
ISSN : 2641-7707
Launched : 2017
Journal of Preventive Medicine and Health Care
ISSN : 2576-0084
Launched : 2018
Journal of Chronic Diseases and Management
ISSN : 2573-1300
Launched : 2016
Annals of Vaccines and Immunization
ISSN : 2378-9379
Launched : 2014
JSM Heart Surgery Cases and Images
ISSN : 2578-3157
Launched : 2016
Annals of Reproductive Medicine and Treatment
ISSN : 2573-1092
Launched : 2016
JSM Brain Science
ISSN : 2573-1289
Launched : 2016
JSM Biomarkers
ISSN : 2578-3815
Launched : 2014
JSM Biology
ISSN : 2475-9392
Launched : 2016
Archives of Stem Cell and Research
ISSN : 2578-3580
Launched : 2014
Annals of Clinical and Medical Microbiology
ISSN : 2578-3629
Launched : 2014
JSM Pediatric Surgery
ISSN : 2578-3149
Launched : 2017
Journal of Memory Disorder and Rehabilitation
ISSN : 2578-319X
Launched : 2016
JSM Tropical Medicine and Research
ISSN : 2578-3165
Launched : 2016
JSM Head and Face Medicine
ISSN : 2578-3793
Launched : 2016
JSM Cardiothoracic Surgery
ISSN : 2573-1297
Launched : 2016
JSM Bone and Joint Diseases
ISSN : 2578-3351
Launched : 2017
JSM Bioavailability and Bioequivalence
ISSN : 2641-7812
Launched : 2017
JSM Atherosclerosis
ISSN : 2573-1270
Launched : 2016
Journal of Genitourinary Disorders
ISSN : 2641-7790
Launched : 2017
Journal of Fractures and Sprains
ISSN : 2578-3831
Launched : 2016
Journal of Autism and Epilepsy
ISSN : 2641-7774
Launched : 2016
Annals of Marine Biology and Research
ISSN : 2573-105X
Launched : 2014
JSM Health Education & Primary Health Care
ISSN : 2578-3777
Launched : 2016
JSM Communication Disorders
ISSN : 2578-3807
Launched : 2016
Annals of Musculoskeletal Disorders
ISSN : 2578-3599
Launched : 2016
Annals of Virology and Research
ISSN : 2573-1122
Launched : 2014
JSM Renal Medicine
ISSN : 2573-1637
Launched : 2016
Journal of Muscle Health
ISSN : 2578-3823
Launched : 2016
JSM Genetics and Genomics
ISSN : 2334-1823
Launched : 2013
JSM Anxiety and Depression
ISSN : 2475-9139
Launched : 2016
Clinical Journal of Heart Diseases
ISSN : 2641-7766
Launched : 2016
Annals of Medicinal Chemistry and Research
ISSN : 2378-9336
Launched : 2014
JSM Pain and Management
ISSN : 2578-3378
Launched : 2016
JSM Women's Health
ISSN : 2578-3696
Launched : 2016
Clinical Research in HIV or AIDS
ISSN : 2374-0094
Launched : 2013
Journal of Endocrinology, Diabetes and Obesity
ISSN : 2333-6692
Launched : 2013
Journal of Substance Abuse and Alcoholism
ISSN : 2373-9363
Launched : 2013
JSM Neurosurgery and Spine
ISSN : 2373-9479
Launched : 2013
Journal of Liver and Clinical Research
ISSN : 2379-0830
Launched : 2014
Journal of Drug Design and Research
ISSN : 2379-089X
Launched : 2014
JSM Clinical Oncology and Research
ISSN : 2373-938X
Launched : 2013
JSM Bioinformatics, Genomics and Proteomics
ISSN : 2576-1102
Launched : 2014
JSM Chemistry
ISSN : 2334-1831
Launched : 2013
Journal of Trauma and Care
ISSN : 2573-1246
Launched : 2014
JSM Surgical Oncology and Research
ISSN : 2578-3688
Launched : 2016
Annals of Food Processing and Preservation
ISSN : 2573-1033
Launched : 2016
Journal of Radiology and Radiation Therapy
ISSN : 2333-7095
Launched : 2013
JSM Physical Medicine and Rehabilitation
ISSN : 2578-3572
Launched : 2016
Annals of Clinical Pathology
ISSN : 2373-9282
Launched : 2013
Annals of Cardiovascular Diseases
ISSN : 2641-7731
Launched : 2016
Journal of Behavior
ISSN : 2576-0076
Launched : 2016
Annals of Clinical and Experimental Metabolism
ISSN : 2572-2492
Launched : 2016
Clinical Research in Infectious Diseases
ISSN : 2379-0636
Launched : 2013
JSM Microbiology
ISSN : 2333-6455
Launched : 2013
Journal of Urology and Research
ISSN : 2379-951X
Launched : 2014
Journal of Family Medicine and Community Health
ISSN : 2379-0547
Launched : 2013
Annals of Pregnancy and Care
ISSN : 2578-336X
Launched : 2017
JSM Cell and Developmental Biology
ISSN : 2379-061X
Launched : 2013
Annals of Aquaculture and Research
ISSN : 2379-0881
Launched : 2014
Clinical Research in Pulmonology
ISSN : 2333-6625
Launched : 2013
Journal of Immunology and Clinical Research
ISSN : 2333-6714
Launched : 2013
Annals of Forensic Research and Analysis
ISSN : 2378-9476
Launched : 2014
JSM Biochemistry and Molecular Biology
ISSN : 2333-7109
Launched : 2013
Annals of Breast Cancer Research
ISSN : 2641-7685
Launched : 2016
Annals of Gerontology and Geriatric Research
ISSN : 2378-9409
Launched : 2014
Journal of Sleep Medicine and Disorders
ISSN : 2379-0822
Launched : 2014
JSM Burns and Trauma
ISSN : 2475-9406
Launched : 2016
Chemical Engineering and Process Techniques
ISSN : 2333-6633
Launched : 2013
Annals of Clinical Cytology and Pathology
ISSN : 2475-9430
Launched : 2014
JSM Allergy and Asthma
ISSN : 2573-1254
Launched : 2016
Journal of Neurological Disorders and Stroke
ISSN : 2334-2307
Launched : 2013
Annals of Sports Medicine and Research
ISSN : 2379-0571
Launched : 2014
JSM Sexual Medicine
ISSN : 2578-3718
Launched : 2016
JSM Biotechnology and Biomedical Engineering
ISSN : 2333-7117
Launched : 2013
Journal of Hematology and Transfusion
ISSN : 2333-6684
Launched : 2013
JSM Environmental Science and Ecology
ISSN : 2333-7141
Launched : 2013
Journal of Cardiology and Clinical Research
ISSN : 2333-6676
Launched : 2013
JSM Nanotechnology and Nanomedicine
ISSN : 2334-1815
Launched : 2013
Journal of Ear, Nose and Throat Disorders
ISSN : 2475-9473
Launched : 2016
JSM Ophthalmology
ISSN : 2333-6447
Launched : 2013
Journal of Pharmacology and Clinical Toxicology
ISSN : 2333-7079
Launched : 2013
Annals of Psychiatry and Mental Health
ISSN : 2374-0124
Launched : 2013
Medical Journal of Obstetrics and Gynecology
ISSN : 2333-6439
Launched : 2013
Annals of Pediatrics and Child Health
ISSN : 2373-9312
Launched : 2013
JSM Clinical Pharmaceutics
ISSN : 2379-9498
Launched : 2014
JSM Foot and Ankle
ISSN : 2475-9112
Launched : 2016
JSM Alzheimer's Disease and Related Dementia
ISSN : 2378-9565
Launched : 2014
Journal of Addiction Medicine and Therapy
ISSN : 2333-665X
Launched : 2013
Journal of Veterinary Medicine and Research
ISSN : 2378-931X
Launched : 2013
Annals of Public Health and Research
ISSN : 2378-9328
Launched : 2014
Annals of Orthopedics and Rheumatology
ISSN : 2373-9290
Launched : 2013
Journal of Clinical Nephrology and Research
ISSN : 2379-0652
Launched : 2014
Annals of Community Medicine and Practice
ISSN : 2475-9465
Launched : 2014
Annals of Biometrics and Biostatistics
ISSN : 2374-0116
Launched : 2013
JSM Clinical Case Reports
ISSN : 2373-9819
Launched : 2013
Journal of Cancer Biology and Research
ISSN : 2373-9436
Launched : 2013
Journal of Surgery and Transplantation Science
ISSN : 2379-0911
Launched : 2013
Journal of Dermatology and Clinical Research
ISSN : 2373-9371
Launched : 2013
JSM Gastroenterology and Hepatology
ISSN : 2373-9487
Launched : 2013
Annals of Nursing and Practice
ISSN : 2379-9501
Launched : 2014
JSM Dentistry
ISSN : 2333-7133
Launched : 2013
Author Information X