Home
JournalsCollections
For Authors For Reviewers For Editorial Board Members
Article Processing Charges Open Access
Ethics Advertising Policy
Editorial Policy Resource Center
Company Information Contact Us
OPEN ACCESS

Emerging Roles of High-mobility Group Box-1 in Liver Disease

  • Lu Wang1,#,
  • Zhiwei Dong2,#,
  • Yeqiong Zhang3,* and
  • Liang Peng3,* 
Journal of Clinical and Translational Hepatology   2024

doi: 10.14218/JCTH.2024.00317

Received:

Revised:

Accepted:

Published online:

 Author information

Citation: Wang L, Dong Z, Zhang Y, Peng L. Emerging Roles of High-mobility Group Box-1 in Liver Disease. J Clin Transl Hepatol. Published online: Oct 22, 2024. doi: 10.14218/JCTH.2024.00317.

Abstract

High-mobility group box-1 (HMGB1) is an architectural chromosomal protein with various roles depending on its cellular localization. Extracellular HMGB1 functions as a prototypical damage-associated molecular pattern that triggers inflammation and adaptive immune responses, mediated by specific cell surface receptors, including receptors for advanced glycation end products and toll-like receptors. Post-translational modifications of HMGB1 significantly impact various cellular processes that contribute to the pathogenesis of liver diseases. Recent studies have highlighted the close relationship between HMGB1 and the pathogenesis of acute liver injuries, including acetaminophen-induced liver injury, hepatic ischemia-reperfusion injury, and acute liver failure. In chronic liver diseases, HMGB1 plays a role in nonalcoholic fatty liver disease, alcohol-associated liver disease, liver fibrosis, and hepatocellular carcinoma. Targeting HMGB1 as a therapeutic approach, either by inhibiting its release or blocking its extracellular function, is a promising strategy for treating liver diseases. This review aimed to summarize the available evidence on HMGB1’s role in liver disease, focusing on its multifaceted signaling pathways, impact on disease progression, and the translation of these findings into clinical interventions.

Graphical Abstract

Keywords

High-mobility group box-1, Liver disease, Damage-associated molecular pattern, Pathogenesis, Signaling pathway, Post-translational modifications

Introduction

Acute liver injury (ALI) is a critical health concern with high morbidity and mortality, commonly triggered by bacterial endotoxin/lipopolysaccharide (LPS) and drug overdoses, such as acetaminophen (APAP).1,2 Clinical treatment options for APAP-induced liver injury are currently limited, and the condition can rapidly progress to acute liver failure (ALF).3 Nonalcoholic fatty liver disease (NAFLD) and alcohol-associated liver disease (ALD) are primary contributors to the prevalence of chronic liver disease (CLD) in Western countries. The global incidence of these conditions has steadily increased in recent years, largely due to shifts in lifestyle and dietary patterns.4,5 CLD can lead to critical complications, such as cirrhosis and hepatocellular carcinoma (HCC), which carry a poor prognosis in advanced stages. Consequently, identifying biological markers that facilitate early diagnosis of ALI and CLD is crucial.

High-mobility group box-1 (HMGB1), the most abundant non-histone nuclear protein, plays a major role in regulating DNA structure by binding to and bending DNA through the minor groove.6 Recent studies have highlighted a notable increase in circulating serum HMGB1 levels in certain CLDs and ALI, indicating a close association between HMGB1 and these conditions.7 Therefore, targeted interventions focused on HMGB1, such as inhibiting its synthesis and release or disrupting its signaling pathways through its receptors, hold promise for mitigating disease progression.8 Moreover, emerging evidence suggests that HMGB1 is a mediator of liver diseases and a valuable biomarker for diagnosis and prognosis.9–11 Compared to previous studies, this review provides a more comprehensive overview of the key types of HMGB1 post-translational modifications (PTMs) in liver disease, with a particular focus on the novel modification of lactylation. Furthermore, it updates recent findings on new molecular mechanisms and therapeutic targets associated with HMGB1 in various liver conditions, offering more cutting-edge insights. Thus, this review synthesizes current literature to elucidate the multifaceted roles and signaling pathways of HMGB1 in CLDs and ALI.

HMGB1

HMGB1 has multiple biological functions depending on its location. It is primarily translocated into the nucleus for architectural functions, including cell cycle regulation, cell death, and DNA replication/remodeling/repair.12 Extracellular HMGB1 functions as a prototypical damage-associated molecular pattern (DAMP) that triggers inflammation and adaptive immune responses.13 Receptors for advanced glycation end products (RAGE) and toll-like receptor (TLR) 4 are among the most prevalent and well-studied extracellular HMGB1 receptors.14

HMGB1 structure

HMGB1 expression is ubiquitous and conserved in higher eukaryotic species.15 HMGB1 contains 215 residues organized into three domains: two DNA-binding regions [an A box (aa 9–79) and a B box (aa 95–163)], a C-terminus, and an N-terminus (Fig. 1).16 The N-terminus of HMGB1 contains many positively charged lysine residues, while the C-terminus is rich in aspartate and glutamate.17 The A box reportedly has anti-inflammatory activity, whereas the B box exhibits pro-inflammatory effects as a specialized antagonist. HMGB1’s steady nuclear localization is mediated by two nuclear localization signals (NLSs), NLS1 (aa 28–44) and NLS2 (aa 179–185).18 These NLSs contain specific sites for PTMs, such as oxidation (aa 23, aa 45, aa 105), acetylation (aa 28–30, aa 180, aa 181–185), and phosphorylation (aa 35), in liver disease. In subsequent sections, we will discuss extensively the impact of HMGB1 PTMs on its function and its crucial role in liver disease. Additionally, HMGB1 has two essential regions for receptor binding, aa 89–108 (TLR4) and aa 150–183 (RAGE).19

Structure of HMGB1 protein.
Fig. 1  Structure of HMGB1 protein.

HMGB1 consists of three domains: two DNA-binding domains (A box and B box) and an acidic tail. The binding sites for TLR4 and RAGE on HMGB1 are specifically located at positions aa 89–108 and 150–183, respectively. NLSs serve as the primary sites for HMGB1 post-translational modifications. Cysteines can be oxidized at C23, C45, and C106. Lysines can undergo acetylation at K28-30, K180, and K181-185. A serine is subject to phosphorylation at S35. However, the specific modification sites for lactylation remain unclear. HMGB1, high-mobility group box-1; NLSs, nuclear localization signals; RAGE, receptor for advanced glycation end products; TLR, toll-like receptor; Ox, oxidation; P, phosphorylation; Ac, acetylation; Lac, lactylation.

HMGB1 functions

HMGB1 performs various functions depending on its cellular localization. Within the nucleus, HMGB1 acts as a DNA chaperone, preserving chromosomal structure and function by maintaining nucleosome integrity, transcription, and DNA repair.20 HMGB1 is passively released by necrotic cells or actively secreted by immune cells, making it a crucial mediator of inflammation, cell migration, and cell proliferation. Extracellular HMGB1 uniquely functions as a prototypical DAMP, acting as a standard alarm.21 It triggers innate immune responses independently or through interaction with cytokines and other molecules by binding to various receptors.22 Hence, HMGB1 is frequently implicated in various pathological conditions, including sepsis, arthritis, cancer, and neurodegenerative diseases. Additionally, HMGB1 has been involved in tissue regeneration. Extracellular HMGB1 can stimulate the proliferation and migration of stem cells, contributing to tissue repair and regeneration.23,24 Moreover, HMGB1 plays a crucial role in regulating autophagy through various intracellular and extracellular signaling pathways.25,26 A comprehensive understanding of HMGB1’s multifaceted roles is essential, as dysregulation of its expression or function can cause pathological effects.

HMGB1 receptors

HMGB1 modulates inflammatory signaling cascades by binding to various receptors, including RAGE, TLR2/4/9, CXCL12/CXCR4, Mac-1, syndecan-1, and differentiation clusters.27–29 We focus on RAGE and TLR4 due to their significance in liver disease.30

TLR4 is a key receptor involved in HMGB1 recognition and is released during liver injury and inflammation. It is expressed in various liver cells, including hepatocytes, Kupffer cells (KCs), endothelial cells, and hepatic stellate cells (HSCs).31 TLR4 activation by extracellular HMGB1 initiates downstream signaling pathways, notably the nuclear factor-kappa B (NF-κB) pathway, which induces the production of pro-inflammatory cytokines and chemokines in hepatocytes.32 This cascade amplifies the immune response in the liver and contributes to tissue damage and inflammation. The HMGB1/TLR4 signaling pathway has been implicated in the pathogenesis of various liver diseases, including NAFLD, ALD, viral hepatitis, and liver fibrosis.33–35 The activation of this inflammatory signaling pathway is closely associated with liver disease progression. Therefore, targeting TLR4 and its associated signaling pathways in HMGB1-mediated liver disease has emerged as a promising therapeutic strategy.

RAGE is a transmembrane receptor belonging to the immunoglobulin superfamily. The interaction between HMGB1 and RAGE triggers signaling cascades that initiate inflammation, tissue regeneration, and immune responses.36 Recent studies have provided valuable insights into the role of RAGE in liver disease.37–39 It is expressed in hepatocytes, HSCs, liver sinusoidal endothelial cells (LSECs), KCs, and oval cells. Liver tissues collected from patients with ALD, NAFLD, and liver fibrosis exhibit elevated RAGE expression. Increased RAGE levels have been associated with the severity and progression of these diseases, suggesting a potential role for RAGE in liver pathology.40,41 RAGE activation triggers downstream signaling cascades, including mitogen-activated protein kinases (MAPKs) and NF-κB pathways in hepatocytes,42 promoting hepatic inflammation. HMGB1-RAGE interactions exacerbate immune cell recruitment to the liver, amplifying inflammatory responses. Additionally, the HMGB1-RAGE axis is crucial in liver fibrosis. The binding of RAGE to HMGB1 activates HSCs, which are vital effector cells in liver fibrosis. HSC activation induces the production of extracellular matrix proteins, causing fibrotic scar formation and tissue remodeling.37 Inhibition of the HMGB1-RAGE axis has shown promise for attenuating liver fibrosis in experimental models. RAGE is a crucial receptor for HMGB1, which regulates HCC proliferation and metastasis.39 Conversely, modulation of the HMGB1-RAGE axis has been associated with enhanced liver regeneration and repair, emphasizing the dynamic role of this interaction in liver homeostasis.

PTMs of HMGB1 in liver disease

PTMs are crucial regulatory mechanisms for cellular proteins, serving various biological functions and modifying the charge state, hydrophobicity, conformation, and stability of proteins.43,44 HMGB1, in particular, undergoes various PTMs.45–51 Specific HMGB1 modifications significantly impact various cellular processes, including DNA stability, transcriptional regulation, protein localization, cell motility, and pro-inflammatory or pro-fibrogenic effects.52–54 Moreover, several crucial HMGB1 PTMs, such as oxidation, phosphorylation, and acetylation, have been identified as contributors to the pathogenesis of liver diseases. Notably, lactylation has been investigated as a mediator of HMGB1 secretion from hepatocytes following hepatic ischemia-reperfusion injury (HIRI).55 The following section focuses on the vital modifications involved in regulating liver diseases and the latest research advancements (Table 1).55–68

Table 1

Functions and therapeutic strategies of HMGB1 PTMs in various liver disease

PTMsLiver diseaseFunctionTherapeutic strategies
AcetylationALD
ALF
HIRI
Regulates the nucleus-cytoplasm shuttling of HMGB1SIRT1 alleviates HMGB1 acetylation and translocation, ameliorating ALD56
SphK1 inhibition diminishes HMGB1 intracellular translocation in ALF57
Pachymic acid alleviates hepatic injury via SIRT1/HMGB1 signal pathway58
PhosphorylationALD
ALI
Sepsis
Promotes HMGB1 secretion to cytoplasm58-F or caspase-11/GsdmD inhibition protects against liver injury5961
OxidationLiver fibrosis
Hepatitis
HCC
Induces translocation of HMGB1 from the nucleus to the cytoplasmNilotinib can improve liver fibrosis via RAGE/HMGB1 axis62
HBx induces HMGB1 oxidation and NLRP3 activation, mediating liver inflammation63
HMGB1 oxidation modulates the proliferation, migration, and metastasis abilities of tumor cells in HCC6467
LactylationHIRI
Sepsis
Increases HMGB1 cytoplasmic accumu-lation in hepatocytes or macrophagesHSPA12A reduces macrophage inflammation by inhibiting lactate production, decreasing HMGB1 lactylation and hepatocyte exosome secretion55
Inhibiting intracellular lactate or blocking lactate signaling reduces HMGB1 lactylation, improving polymicrobial sepsis68

Acetylation of HMGB1 in liver disease

The acetylation of specific lysine residues in HMGB1 has been associated with its ability to modulate inflammatory responses, immune functions, and apoptosis.69 Multiple lysine acetylation sites have been identified in NLS1 and NLS2. HMGB1 acetylation reportedly contributes to liver inflammation and injury in ALD by affecting its subcellular localization, DNA-binding activity, and interactions with other proteins.70 HMGB1 acetylation has been shown to facilitate its translocation from the nucleus to the cytoplasm in cell and mouse models of alcohol-induced liver injury.56 This pro-inflammatory effect was suppressed following treatment with resveratrol, a potent agonist of sirtuin 1 (SIRT1), by inhibiting HMGB1 acetylation and translocation. Moreover, HMGB1 acetylation is crucial in the pathogenesis of sepsis-associated liver injury. LPS induces HMGB1 expression in KCs and regulates its acetylation, influencing its intracellular translocation. Tian et al. found that sphingosine kinase 1 (SphK1) inhibition markedly improves sepsis-associated liver injury by inhibiting HMGB1 expression, intracellular translocation, and acetylation.57 Additionally, HMGB1 acetylation exacerbates oxygen-glucose deprivation/reperfusion injury by promoting its translocation and release, leading to increased pro-inflammatory cytokine levels, oxidative stress, and hepatocellular apoptosis.58

Phosphorylation of HMGB1 in liver disease

The phosphorylation of serine residues within the NLSs of HMGB1 regulates its nucleocytoplasmic translocation, which is a critical step in its release into the extracellular space.59 Zhao et al. found that HMGB1 phosphorylation promotes its release from the nucleus to the cytoplasm, mediating lipid degeneration in hepatocytes and facilitating the progression of ALD.71 Calcium dysregulation during liver injury causes an elevated intracellular calcium load in hepatocytes.72 The translocation and release of HMGB1 are reportedly regulated by the calcium/calcium-dependent kinase signaling pathway, which serves as an upstream signaling mechanism for HMGB1 phosphorylation. Activation of calcium-dependent kinases, such as PKCα and calmodulin-dependent protein kinase (CaMK) IV, promotes HMGB1 phosphorylation in H2O2-induced liver injury, facilitating its cytoplasmic translocation, subsequent release, and immuno-regulatory effect.60 Similarly, Li et al. found that HMGB1 phosphorylation mediated by protein kinase C or calcium/CaMKII was associated with its release from hepatocytes in response to LPS stimulation.61 However, the specific phosphorylation sites within HMGB1-NLS remain unclear.

Oxidation of HMGB1 in liver disease

HMGB1 oxidation primarily affects cysteine, lysine, and tyrosine residues, leading to their modification by reactive oxygen species (ROS), such as superoxide anions, hydrogen peroxide, and other oxidants, such as peroxynitrite.48 HMGB1 oxidation can induce structural alterations that influence its protein-protein interactions, DNA-binding capacity, and overall functionality in diverse biological processes. Investigations into HMGB1 oxidation have provided significant insights into its involvement in several diseases, such as liver fibrosis, HCC, and hepatitis. Oxidative stress is crucial in HSC activation during liver injury progression.73 HMGB1 oxidation reportedly regulates liver fibrosis by activating HSCs and promoting collagen synthesis via RAGE/HMGB1/TGF-β and oxidative stress pathways.62 Moreover, hepatitis B virus (HBV)-encoded X protein triggers HMGB1 oxidation and extracellular release in H2O2-stimulated HL7702 cells. The oxidized HMGB1 activates the NLRP3 inflammasome, inducing liver tissue inflammation and hepatocyte pyroptosis.63 Thus, oxidized HMGB1 acts as a signaling molecule to regulate inflammatory reactions in HBV-infected tissues. Recent studies have shown that HMGB1 oxidation significantly influences HCC by modulating the proliferation and migration of tumor cells.64 Notably, HMGB1-induced metastasis of tumor cells is considered an inflammatory response triggered by hypoxia stress through the RAGE/NF-κB and TLR4/caspase-1 signaling pathways.75,66 Jing et al. proposed a novel mechanism by which HMGB1 oxidation promotes HCC proliferation and metastasis. HMGB1 upregulates the expression of the mitochondrial transport protein RHOT1 and tunneling nanotube-related protein RAC1 in hypoxic environments, promoting mitochondrial transfer and infiltration of tumor cells.67 This novel mechanism provides a new perspective for exploring therapeutic strategies for HCC.

Lactylation of HMGB1 in liver disease

Lactylation is a novel epigenetic modification, heralding a new era for the comprehensive investigation of lactate metabolism.74,75 Lactylation modifications alter protein structure and function, influencing protein folding, stability, and hydrophobicity. Consequently, they affect crucial biological processes, such as cellular signal transduction, cell cycle regulation, and metabolic control.76 Multiple studies have demonstrated the essential contribution of protein lactylation in inflammation, cancer, and neuropsychiatric disorders.77,78 Notably, lactylation modification is emerging as a crucial factor in liver disease and is closely associated with the development and progression of various conditions, such as NAFLD, fibrosis, and HCC.79–81 Yang et al. revealed that macrophages can take up lactate through monocarboxylate transporters and facilitate HMGB1 lactylation via the p300/CBP pathway. Inhibiting intracellular lactate levels or blocking lactate signaling pathways can decrease the lactylation level of HMGB1 in exosomes, improving polymicrobial sepsis.68 Furthermore, elevated levels of HMGB1 lactylation in hepatocytes during HIRI were closely associated with macrophage chemotaxis and inflammatory cytokine activation. However, hepatocyte heat shock protein A12A (HSPA12A) mitigates these effects by inhibiting lactate production from glycolysis, reducing HMGB1 lactylation and hepatocyte exosome secretion.55 The Warburg effect is a crucial event in HCC development, where tumor tissues utilize the abundant lactate produced through aerobic glycolysis as a lactylation substrate.82 The lactylation of histones and non-histones has garnered widespread attention, serving as a cornerstone of the tumor microenvironment (TME) and participating in HCC progression by regulating gene expression and cellular metabolism.83–85 Unfortunately, there are currently no relevant publications on the involvement of HMGB1 in the pathogenesis of HCC. The author proposes that, influenced by the Warburg effect, HMGB1—functioning as a classic DAMP—could be a valuable target for elucidating the development and treatment of HCC through its lactylation modifications. Therefore, lactylation serves as a bridge between epigenetic and metabolic reprogramming in cancer cells. These findings suggest that HMGB1 lactylation is crucial in modulating inflammatory responses in liver diseases. However, studies on HMGB1 lactylation are currently limited to polymicrobial sepsis and HIRI. Interestingly, Gao et al. recently confirmed that mitochondrial pyruvate carrier 1 regulates fatty acid synthase lactylation at the K673 site and plays a role in treating NAFLD.80 This is a valuable finding, providing a robust foundation for investigating the role of HMGB1 lactylation in NAFLD. Further in-depth investigations are urgently needed to explore the molecular mechanisms, including acetoacetyl coenzyme A-related writers and erasers, modification sites, and reaction kinetics, to develop new therapeutic interventions for liver disease.

HMGB1 in acute liver disease

HMGB1 in APAP-induced ALI

Acetaminophen-induced acute liver injury (AILI) has emerged as a significant public health concern, ranking among the top causes of drug-induced liver injury in developed nations.86,87 APAP overdose triggers an inflammatory response and oxidative stress in hepatocytes at the onset of AILI. HMGB1, a DAMP associated with AILI, is passively released from necrotic hepatocytes, leading to elevated levels of HMGB1 in the systemic circulation.88–90 Clinical studies have indicated that HMGB1 is a more sensitive biomarker than ALT in predicting the development of liver injury in patients with AILI. Additionally, it is vital in the stratification of early liver damage risk among AILI patients, enabling the customization of treatment strategies in clinical practice.90 Pirnie et al. utilized high-resolution mass spectrometry to identify oxidized modifications of cysteine residues (Cys-23, Cys-45, and Cys-106) in HMGB1, further elucidating the HMGB1 phenotype secreted by hepatocytes in AILI. These findings identify HMGB1 as a potential early biomarker of liver toxicity following APAP overdose.91

HMGB1 enhances the immune response by activating macrophages and recruiting neutrophils during immune-mediated liver injury. This results in the release of excess cytotoxic ROS and proteases, exacerbating liver damage.92 Extracellular HMGB1 binds to the RAGE receptor on the surface of neutrophils, selectively inducing their infiltration into necrotic sites, though it does not affect liver macrophage levels.93 Additionally, HMGB1 can activate macrophages via the TLR4 signaling pathway, promoting the secretion of pro-inflammatory mediators, such as interleukin (IL)-23 and IL-17, which induce neutrophil migration and exacerbate liver damage.94 HMGB1 also plays a crucial role in activating the macrophage NLRP3 inflammasome during AILI.1 Activated NLRP3 inflammasomes mediate neutrophil infiltration and hepatocyte apoptosis by inducing Caspase-1 activation and upregulating IL-1β expression.95 In the development of AILI, HMGB1 interacts with the TLR4 receptor and triggers macrophage activation through CD36, mediating Erk and Akt signaling and upregulating IL-1β and IL-6 expression. However, further investigation is needed to understand how HMGB1 mediates the function of CD36.96 These studies substantiated the significance of HMGB1 in the innate immune modulation of AILI, facilitating crosstalk between hepatocytes and neutrophils. According to reports, extracellular HMGB1 can stimulate immune cells and trigger the necrosis of adjacent liver cells via the TLR4/TRIF/RIPK3 signaling pathway, accelerating liver cell necrosis.97 Liu et al. proposed a mechanism in which APAP-induced activation of Caspase-1 in hepatocytes leads to neutrophil extracellular trap generation and subsequent release of pro-inflammatory cytokines. Notably, the depletion or inhibition of neutrophil extracellular traps in neutrophils reduces HMGB1 levels and prevents hepatocyte necrosis.98

N-acetylcysteine is currently the most effective treatment for APAP-induced liver damage; however, its therapeutic window is limited. Consequently, alternative therapies for treating AILI are required.99 Treatment with a partially humanized anti-HMGB1 monoclonal antibody has shown higher efficacy and a broader therapeutic window in AILI compared to N-acetylcysteine.100 Moreover, HMGB1-neutralizing antibodies have been shown to effectively decrease serum HMGB1 levels and bacterial translocation.101 Additionally, sesamin exhibits a protective effect against AILI by mitigating hepatic oxidative stress and inflammatory responses through the inhibition of the HMGB1/TLR4/NF-κB signaling pathway, leading to decreased hepatocyte apoptosis levels.102 Therefore, HMGB1 may serve as a pivotal therapeutic target for preventing APAP overdose-induced liver damage.

HMGB1 in HIRI

HIRI is a common complication of hepatic resection and liver transplantation.103 The pathological progression of HIRI involves two interconnected phases: the onset of localized ischemia leading to hepatocyte injury, and the subsequent release of DAMPs, which triggers immune cell activation and initiates an inflammatory cascade that exacerbates liver damage.104,105 HMGB1 serves as a crucial alarm molecule during the early stages of HIRI.106 Numerous preclinical and clinical studies have confirmed that inhibiting HMGB1 release can effectively mitigate liver injury.107,108 The release of serum HMGB1 after HIRI primarily depends on TLR4 activation.109 TLR4 can specifically identify hepatocytes and induce HMGB1 release by activating the phosphorylation of the c-Jun N-terminal kinase and p38 signaling pathways. Therefore, hepatocytes are the primary contributors to the circulatory release of HMGB1 during HIRI.110,111 Du et al. identified a novel regulatory mechanism for HMGB1 secretion by hepatocytes.55 HMGB1 lactylation promotes its extracellular translocation from the nucleus, orchestrating macrophage chemotaxis and inflammatory activation. HSPA12A exerts strong hepatoprotective effects by inhibiting lactate production through glycolysis, reducing HMGB1 lactylation and secretion (Fig. 2). This study establishes a strong link between lactylation and HMGB1 secretion, highlighting its potential as a future therapeutic target for HIRI.

The mechanism of HMGB1 release and its pathological impact in HIRI.
Fig. 2  The mechanism of HMGB1 release and its pathological impact in HIRI.

HMGB1 lactylation promotes its extracellular translocation from the nucleus. HSPA12A can inhibit lactate production from glycolysis, thereby reducing HMGB1 lactylation and secretion. Extracellular HMGB1 binds to specific receptors on KCs, initiating an innate immune response. Moreover, HMGB1 specifically targets LSECs, leading to the nuclear translocation of IRF1 and CXCL1, which triggers neutrophil recruitment. ↑, increase; ↓, decrease; HMGB1, high-mobility group box-1; HIRI, hepatic ischemia-reperfusion injury; HSPA12A, heat shock protein A12A; pJNK, phosphorylation of c-Jun N-terminal kinase; LSECs, liver sinusoidal endothelial cells; NE, neutrophils; RAGE, receptor for advanced glycation end products; ROS, reactive oxygen species; TLR, toll-like receptor.

Notably, in early-stage HIRI, the activation of TLR4 on KCs triggers an innate immune response before neutrophil recruitment from the peripheral blood to the injured liver. The HMGB1/TLR4/NF-κB signaling pathway plays a critical role in the inflammatory cascade, leading to secondary immune injury following hepatic hypoxic.112 Recent studies have focused on this signaling pathway and investigated the protective effects of various compounds, including drugs,113,114 γ-oryzanol,115 bioactive peptides (Ac2-26),116 and TLR4 antagonists (eritoran),117 which possess anti-inflammatory, antioxidant, and anti-apoptotic properties in HIRI. Additionally, RAGE is involved in HMGB1-mediated sterile inflammatory cascades.118

HMGB1 is a crucial mediator between hepatocytes and other liver cells, influencing the immune microenvironment and contributing to HIRI. LSECs play vital protective roles by maintaining vascular homeostasis, controlling inflammation, regulating vascular tone, and facilitating toxin clearance.119 (Fig. 2) Upon recognition by TLR3/TLR4, extracellular HMGB1 specifically targets LSECs, leading to the nuclear translocation of IRF1 and subsequent transcription of CXCL1. This molecular cascade triggers neutrophil chemotaxis and accelerates the senescence-associated secretory phenotype in LSECs, further disrupting the liver sinusoid-like immune microenvironment. However, acteoside has shown protective effects against HIRI by targeting the HMGB1-TLR3/4-IRF1 signaling pathway and reversing LSEC senescence.120 Tanemura et al. demonstrated that dabigatran enhances the expression of endogenous thrombomodulin in LSECs, reducing excessive serum HMGB1 levels, alleviating inflammation and cell apoptosis, preserving vascular integrity, and mitigating ischemia/reperfusion liver injury.121

HMGB1 in ALF

ALF is a prevalent and critical clinical condition stemming from various factors, including APAP toxicity, viral and autoimmune hepatitis, hepatic ischemia, drug-induced liver injury, and the use of herbal supplements. During the initial stage of ALF, the predominant pathogenic processes typically involve widespread hepatocyte necrosis and apoptosis within the liver tissue.122 The release and activation of HMGB1 are crucial in the pro-inflammatory network that characterizes the pathophysiology of ALF.123,124 Dynamic network analyses have revealed that serum HMGB1 expression is significantly higher than other inflammatory mediators in pediatric patients with ALF, suggesting that HMGB1 can serve as a critical predictive indicator for survival outcomes in affected children.11 Furthermore, clinical cohort studies have confirmed that serum HMGB1 is an early and reliable biomarker for predicting acute kidney injury in patients with HBV-related acute-on-chronic liver failure, which is closely associated with poor prognosis.125

Hepatocyte death is recognized as a momentous event and a significant contributor to the progression of ALF. Notably, HMGB1 is significant in mediating various forms of cell death following immune imbalance in ALF.126 For example, the tumor necrosis factor (TNF)-α/HMGB1 signaling pathway plays a pivotal role in triggering hepatocyte necroptosis in LPS/D-galactosamine-induced ALF. Treatment with TNF-α inhibitors (CC-5013) or glycyrrhizin can effectively inhibit HMGB1 release, ameliorating hepatocyte necroptosis and liver tissue damage.127 Similarly, Wang et al. discovered that macrophages actively release extracellular vesicles (EVs) loaded with HMGB1, which selectively target hepatocytes. Through the HMGB1/RAGE signaling pathway, HMGB1 induces NLRP3 inflammasome activation, leading to hepatocyte necroptosis.128 Furthermore, the levels of HMGB1-containing EVs are closely correlated with the degree of liver tissue injury. This highlights the significance of HMGB1-containing EVs as mediators of the communication between macrophages and hepatocytes, offering a promising strategy for targeted therapeutic interventions. Moreover, HMGB1 is crucial in mediating ferroptosis in ALF. The HMGB1 inhibitor glycyrrhizin has demonstrated a notable anti-ferroptotic effect in ALF by effectively decreasing Fe2+ and ROS levels while increasing glutathione levels in liver tissue.129 However, the precise molecular mechanisms underlying HMGB1-mediated ferroptosis remain unclear and warrant further investigation.

Studies have shown that inhibiting HMGB1 activity or release can effectively mitigate the progression of ALF. The HMGB1-A box effectively suppresses the inflammatory activity of HMGB1 through competitive binding in LPS/D-Gal-induced ALF. The HMGB1-A box inhibits the TLR4/NF-κB signaling pathway, thereby reducing hepatocyte apoptosis and pro-inflammatory cytokine levels.124 Additionally, the anti-inflammatory agent heparan sulfate selectively binds to HMGB1, targeting the HMGB1/RAGE axis and demonstrating effective hepatoprotective effects in APAP-induced ALF.130 Furthermore, histone deacetyltransferase 4 (hereinafter referred to as HDAC4) is a vital regulator involved in the acetylation and nucleocytoplasmic shuttling of HMGB1. Tian et al. demonstrated that the SphK1/CaMKII-δ pathway serves as an upstream regulator of HDAC4 phosphorylation. Thus, SphK1 inhibition significantly reduces intracellular HMGB1 release, providing a potential approach for treating sepsis-related liver injury.57

HMGB1 in CLD

HMGB1 in NAFLD

The NAFLD incidence has risen significantly, transforming it into a global public health concern.131 As the condition progresses, approximately 30% of patients with NAFLD develop an inflammatory state in the liver known as nonalcoholic steatohepatitis (NASH).132 HMGB1 is an early mediator in the progression of NASH and is implicated in the pro-inflammatory microenvironment created by lipotoxicity.33 Two clinical investigations were conducted to further investigate the potential role of HMGB1 as an early diagnostic biomarker of NAFLD. However, findings regarding the association between HMGB1 levels and the degree of liver fibrosis are inconclusive. One study reported a robust correlation between serum HMGB1 levels and liver fibrosis in pediatric patients with NAFLD.7 Conversely, another study revealed no significant relationship between HMGB1 levels and liver inflammation in either pediatric or adult patients with NAFLD.133 This discrepancy may be owing to the varied degrees of liver fibrosis among these patients. Additionally, no direct correlation between peripheral HMGB1 levels and the degree of liver tissue fibrosis has been established, highlighting the need for further research in this area. Sarcopenia is a severe condition common to various CLDs,134,135 and timely identification and precise evaluation are essential. Recent studies have shown that sarcopenia is closely associated with NAFLD and serves as an independent risk factor for the progression of the disease.136 Notably, studies have confirmed that HMGB1 plays a vital role in the pathogenesis of sarcopenia.137,138 HMGB1 promotes skeletal muscle atrophy through an IL-18-dependent mechanism.139 The elevation of IL-18 production induced by HMGB1 is mediated by the RAGE/p85/Akt/mTOR/c-Jun signaling pathway, indicating that the HMGB1/IL-18 pathway is a promising target for treating sarcopenia.

HMGB1 plays a dual role in the pathogenesis of NAFLD, potentially facilitating the progression of NASH or ameliorating hepatic lipotoxic injury, depending on its precise cellular localization. The upregulation of HMGB1 expression in hepatocytes is regulated by the JNK1/2-ATF2 axis and the miR-200 family in response to lipotoxic injury.140 Extracellular HMGB1 initiates the TLR4/MyD88 signaling pathway in hepatocytes and induces an inflammatory response in the infusion of free fatty acids (FFA) or a high-fat diet.33 (Fig. 3) Additionally, extracellular HMGB1 stimulates the expression of inflammatory mediators (TNF-α and IL-6) via the RAGE/p-JNK/p-ERK signaling axis, exacerbating hepatic inflammation and insulin resistance.141 Therefore, extracellular HMGB1 serves as a DAMP and regulates innate immune signaling pathways, promoting the transition from NAFLD to NASH. Suppression of HMGB1 release from hepatocytes or treatment with HMGB1-neutralizing antibodies can mitigate liver damage.142 Conversely, the intracellular function of HMGB1 in the development of NAFLD has emerged as a major focus of recent studies. Lin et al. demonstrated that hepatocyte-specific HMGB1 (HC-HMGB1) is crucial in maintaining endoplasmic reticulum (ER) homeostasis and preventing FFA-induced liver damage (Fig. 3). Compared with wild-type mice, HC-HMGB1-knockout (−/−) mice exhibited reduced expression of β-oxidation genes and increased ER stress markers in the liver, resulting in increased lipid accumulation and cell injury.143 Nuclear HMGB1 is a potent inhibitor of liver lipogenesis. By specifically deleting the HMGB1 gene in hepatocytes, nuclear HMGB1 effectively inhibits the activities of the LXRα/PPARγ axis, preventing the development of liver steatosis (Fig. 3).144 This finding suggests that HMGB1 might detach from chromatin in response to signals from the microenvironment, potentially influencing metabolic processes. Moreover, a novel HMGB1-mediated autophagy pathway has been reported to be involved in p53 regulation in NAFLD.145 Functional silencing of p53 promotes the translocation of HMGB1 from the nucleus to the cytoplasm. Once in the cytoplasm, HMGB1 triggers autophagy in hepatocytes by activating Beclin-1, facilitating lipid degradation. Although the precise mechanisms by which hepatocellular HMGB1 regulates ER stress and lipid degradation remain unclear, these studies offer innovative insights into potential strategies for preventing the onset of NAFLD. These findings provide new insights for the clinical treatment of NAFLD, suggesting that HMGB1 acts as an alarming signal and plays a pivotal role in lipid metabolism.

Roles of nuclear and extracellular HMGB1 in the pathogenesis of NAFLD.
Fig. 3  Roles of nuclear and extracellular HMGB1 in the pathogenesis of NAFLD.

Functional silencing of p53 promotes HMGB1 nucleocytoplasmic translocation. Then, HMGB1 triggers autophagy in hepatocytes by activating Beclin-1, facilitating lipid degradation. Nuclear HMGB1 also inhibits the LXRα/PPARγ axis, preventing liver steatosis. HC-HMGB1 can inhibit ER stress and prevent FFA β-oxidation. Conversely, extracellular HMGB1 stimulates the expression of TNF-α and IL-6 via the TLR4/MyD88 and RAGE/p-JNK/p-ERK signaling pathways. ↑, increase; ↓, decrease; BCL1, Beclin-1; ER, endoplasmic reticulum; FFA, free fatty acids; HMGB1, high-mobility group box-1; HC-HMGB1, hepatocyte-specific HMGB1; IL, interleukin; pJNK, phosphorylation of c-Jun N-terminal kinase; NAFLD, non-alcoholic fatty liver disease; NASH, nonalcoholic steatohepatitis; RAGE, receptor for advanced glycation end products; TLR, toll-like receptor; TNF, tumor necrosis factor.

HMGB1 in ALD

HMGB1 has been successfully implicated in ALD through various processes, including oxidative stress, mitochondrial dysfunction, inflammatory responses, disruption of lipid metabolism, and exposure to endotoxins, all of which are consequences of excessive alcohol consumption.146 Clinical studies have confirmed that circulatory HMGB1 levels in patients with ALD are significantly elevated, serving as a biomarker to predict the clinical outcomes of alcoholic steatohepatitis.10 As the disease progresses, liver tissue biopsies of patients with ALD show increased translocation of HMGB1 from the nucleus to the cytoplasm.147 The regulatory mechanisms of HMGB1 release are closely associated with PTMs, including acetylation, phosphorylation, and oxidative modifications. Further research revealed that genetic ablation of HMGB1 protects against alcoholic liver damage in mice, underscoring the deleterious impact of HMGB1 on the progression of ALD.71 Moreover, HMGB1 plays a critical role in reducing ER stress in hepatocytes by promoting liver inflammation and apoptosis, which are crucial factors in the development of alcoholic steatohepatitis.148 HSC activation is reportedly a major contributor to liver fibrosis. Previous studies demonstrated that HMGB1 is released from hepatic parenchymal cells upon ethanol exposure. This release promotes the migration of HSCs and LSECs to the site of liver injury, contributing to the development of liver fibrosis.149 Xie et al. found that inhibiting autophagy could reverse alcohol-induced HSC activation and alleviate alcohol-induced liver injury in ALF mice.150 They also identified the long non-coding RNA XIST as a competitive endogenous RNA for miR-29b, promoting HMGB1 cytoplasmic expression and inducing autophagy and HSC activation.151

Recent studies have investigated the effects of HMGB1 regulators or inhibitors on ALD. SIRT1 reportedly plays a crucial role in the negative regulation of HMGB1 in ALD. The administration of SIRT1 agonists has been shown to effectively suppress HMGB1 acetylation and release in hepatic tissue, thereby ameliorating liver injury associated with ALD.56 BRD4 activates inflammatory responses in ALD via the BRD4/HMGB1 signaling pathway. Salvianolic acid A inhibits this pathway by reducing BRD4 expression and preventing HMGB1 release, showing promise for improving ethanol-induced hepatic inflammation, suggesting salvianolic acid A as a potential treatment for ALD.152 Furthermore, digitoflavone can be used to treat ALD by inhibiting the HMGB1/TLR4 axis, reducing ethanol-induced inflammation, decreasing lipid production, and increasing lipid oxidation, thereby mitigating liver damage.153 However, these findings are limited to animal studies, underscoring the need for clinical trials to evaluate the effectiveness and safety of HMGB1 inhibitors in patients with ALD and provide crucial empirical data for their clinical application.

HMGB1 in liver fibrosis

Liver fibrosis is a multifaceted pathological process significantly influenced by inflammation.154 HMGB1 is a classical pro-inflammatory factor that plays a crucial role in HSC activation and extracellular matrix protein production.155,156 HMGB1 expression in the liver is associated with the stage of fibrosis in individuals with hepatitis, primary biliary cirrhosis, and ALD.38 Moreover, a prospective cohort study has demonstrated a potential correlation between elevated serum HMGB1 levels and decreased survival rates in patients with cirrhosis complicated by acute kidney injury.157 Recent studies have focused on elucidating the specific mechanisms by which HMGB1 contributes to liver fibrosis. HMGB1 interacts with RAGE or TLR4 to activate a cascade of signaling pathways, including NF-κB/p65, MAPK, PI3K/Akt, and pMEK1/2/pERK1/2/pc-Jun. It also stimulates the activation and proliferation of HSCs, promoting the progression of fibrosis.62,158,159 Additionally, HMGB1 can stimulate the generation of pro-inflammatory cytokines (such as IL-6, TNF-α, and IL-1), aggravating intrahepatic inflammation and accelerating fibrosis progression.38,62,160,161

IHMGB1 consists of two boxes: the B box, which promotes inflammation, and the A box, which inhibits TLR signaling and limits inflammatory responses.162,163 Based on the unique structural attributes of HMGB1, studies have revealed a peptide derived from the A box that can reduce fibrotic lesions following liver injury. Furthermore, this HMGB1 peptide promotes macrophage polarization towards an anti-inflammatory profile, offering promising therapeutic benefits for treating liver fibrosis.164 He et al. recently proposed an innovative pathogenic mechanism involving HMGB1 in the progression of HBV-induced liver fibrosis.165 Their findings demonstrate that HMGB1 serves as a critical mediator of macrophage NLRP3 inflammasome activation. Additionally, epigallocatechin-3-gallate has been shown to promote the cytoplasmic autophagic degradation of HMGB1, reducing extracellular HMGB1 levels, inhibiting macrophage inflammatory complex activation, and improving liver tissue damage. However, the molecular mechanisms underlying how epigallocatechin-3-gallate inhibits macrophage NLRP3 activation remain unclear. Future studies should focus on identifying precise targets and methods to improve clinical interventions.

Another highly homologous protein, HMGB2, has been implicated in the pathogenesis of liver fibrosis and cirrhosis. Serum HMGB2 levels are significantly elevated in patients with liver fibrosis and cirrhosis. Genetic ablation of HMGB2 in vivo protects against CCl4-induced liver fibrosis owing to HSC inactivation, suggesting that HMGB2 could be a promising target for preventing the development of liver fibrosis.166

HMGB1 in HCC

The development of HCC is intricately associated with a process involving chronic cell death, inflammation, fibrosis, and repeated repair and regeneration responses.167 HMGB1 is vital in the initiation and advancement of HCC. Multiple clinical studies have revealed that HMGB1 levels in the circulation and tumor tissues of patients with HCC are significantly elevated. Moreover, the increased presence of HMGB1 is strongly associated with tumor size, Edmondson grade, and tumor-node-metastasis staging, underscoring a direct association between elevated HMGB1 expression and unfavorable prognosis.66,168,169 The role of HMGB1 in HCC is multifaceted and paradoxical owing to its distinct intracellular and extracellular localization.

HMGB1 promotes hepatocarcinogenesis

Liver-specific HMGB1 deficiency reduces HCC progression in CLD. Its deletion leads to reduced dendritic cells in tumors, suggesting a pivotal role in linking hepatocyte death to ductular reactions and hepatocarcinogenesis in CLD.170 Athavale et al. reported that mice with impaired Hippo signaling pathways showed significant reductions in YAP activity, inflammation, fibrosis, and HCC burden after HMGB1 deletion. HMGB1-deficient mice displayed severe abnormalities in their intrahepatic bile ducts and developed hyperbilirubinemia. This implies that HMGB1 may contribute to the development of HCC and plays a role in maintaining the normal function of intrahepatic bile ducts under Hippo signaling deficiency.171 Researchers recently proposed a regulatory role for HMGB1 in HBV-related early-stage HCC.172 They suggested that elevated HMGB1 mRNA levels upregulate RICTOR mRNA expression by competitively binding to the miR-200 family. This HMGB1-mediated RNA-RNA crosstalk promotes glutamine metabolism in tumor cells, enhancing their stem-like properties and tumorigenesis via epigenetic modifications. Moreover, this crosstalk may affect the effectiveness of immunotherapy by increasing programmed death-ligand 1 (PD-L1) expression and PD-L1+ exosome activity. These findings provide new insights into potential targets for early-stage HCC treatment with anti-PD-L1 therapy. Cao et al. suggested that liver infection with H. hepaticus may influence the development of precancerous liver lesions primarily by promoting HMGB1 activation and accumulation.

HMGB1 promotes tumor proliferation and metastasis

RAGE is a key receptor for HMGB1, which regulates HCC proliferation and metastasis. The HMGB1/NF-κB signaling pathways, such as NF-κB/Ras-MAPK, NF-κB/STAT, and NF-κB/PI3K/Akt, are involved in modulating the expression of crucial signaling molecules and cytokines closely associated with cancer progression.65,173–176 For example, HMGB1 stimulates increased KLF7 expression in tumor cells via the TLR4/RAGE-PI3K-AKT-NF-κB pathway. This causes the establishment of an HMGB1-KLF7-TLR4/PTK2 positive feedback loop, facilitating the development of HCC.177 Autophagy significantly impacts the aggressive proliferation of tumors. A study revealed that HMGB1 serves as a crucial facilitator driving the advancement of HCC in autophagy-deficient livers.170 HMGB1 is released through paracrine signaling and interacts with RAGE receptors on macrophages or ductal cells, stimulating the growth of liver tumors and modifying the TME.178 In addition to conventional receptors, Pu et al. revealed that valosin-containing protein (VCP), a well-known molecule implicated in tumor metastasis and prognosis, significantly influences the advancement of HCC.179 The interaction between the D1 domain of VCP and the A box of HMGB1 activates the PI3K/AKT/mTOR signaling pathway, enhancing the proliferation, migration, and invasion abilities of tumor cells. This demonstrates the pivotal involvement of HMGB1 in the VCP-driven progression of HCC, underscoring the potential of VCP and HMGB1 as promising therapeutic targets for HCC treatment.

HMGB1 regulates the TME

Alterations in the TME are pivotal in the advancement of HCC,180 with HMGB1 serving a critical role in enhancing the production of inflammatory factors,181 stimulating angiogenesis,182,183 and facilitating immune evasion.184,185 In hypoxic environments, HMGB1 expression is significantly increased in HCC, resulting in the infiltration of macrophages and their repolarization to promote IL-6 expression, which enhances the invasiveness and metastasis of tumor cells. Moreover, extracellular HMGB1 acts as a chemoattractant for leukocytes and a pro-inflammatory cytokine, prompting recruited leukocytes and resident immune cells to release TNF-α, IL-1, IL-6, and other cytokines.181 Additionally, HMGB1 expression is increased by hepatitis B virus-encoded X protein activation, which plays a crucial role in the development of HBV-related liver cancer. HMGB1 modulates the IL-6/STAT3/miR-34a signaling pathway, facilitating epithelial-mesenchymal transition and tumor angiogenesis in HBV-related liver cancer. Therefore, HMGB1 may serve as a potential target for invasion and venous metastasis of HBV-related liver cancers.182 Notably, mitochondrial transfer is a recently identified dynamic phenomenon associated with various human diseases, including cancer and cardiovascular diseases.186 Jing et al. indicated that extracellular HMGB1 promotes mitochondrial transfer to influence the TME and the progression of HCC under hypoxic conditions.67 HMGB1 enhances the expression of RHOT1 by boosting the activity of the NF-Y complex, regulating the recruitment of RAC1 to the cell membrane. This process facilitates mitochondrial transfer and promotes the migration and metastasis of tumor cells. Furthermore, elevated levels of HMGB1, RHOT1, and RAC1 in patients with HCC are associated with shorter overall survival. These findings demonstrate the complex interplay between hypoxia, HMGB1, and mitochondrial transfer during the progression of HCC, highlighting potential targets for therapeutic interventions.

Therapeutic potential of HMGB1

As discussed previously, HMGB1 is vital in the pathogenesis of acute and chronic liver diseases.187 Specific strategies have been employed to target HMGB1 in treating these conditions, including inhibiting its release or impeding the function of extracellular HMGB1.8,188

HMGB1 antagonists effectively block excessive extracellular HMGB1 and alleviate inflammatory liver diseases.189 An anti-HMGB1 monoclonal antibody reportedly protects animals against lethal sepsis-induced ALI by inhibiting HMGB1 endocytosis. These therapeutic approaches show promise for regulating HMGB1-mediated immune activation.190 Furthermore, various chemical molecules and HMGB1-based peptides can bind directly to HMGB1 and inhibit its cytoplasmic translocation. Certain Chinese herbal medicines, such as curcumin, glycyrrhizin, and salvianic acid A, exhibit hepatoprotective effects by inhibiting the extracellular release of HMGB1 to alleviate liver inflammation.152,158,191 Moreover, the HMGB1 peptide synthesized from the A box can drive macrophages toward an anti-inflammatory state, attenuating the advancement of liver fibrosis and showing promise as a therapeutic agent for cirrhosis.164 Similarly, the peptide antagonist P5779 protects against experimental HIRI, APAP-induced liver toxicity, and sepsis lethality through the HMGB1/TLR4 signaling pathway.192 Notably, HMGB1 is a crucial therapeutic target in HCC.193 Multiple basic studies have shown that microRNAs (miR-320a, miR-325, miR-505, and miR-129-2) and long non-coding RNA (MIR22HG) are involved in the pathogenesis of HCC by negatively modulating the post-translational expression of HMGB1 in hepatocytes. This regulation suppresses tumor cell proliferation, invasion, and metastasis.194–197

Furthermore, some HMGB1 antagonists can effectively inhibit the transduction of inflammatory signaling pathways associated with HMGB1, thereby reducing its pro-inflammatory effects.102 Digitoflavone has demonstrated potential in managing ALD by inhibiting the HMGB1-TLR4 signaling pathway.153 In APAP-induced ALF, heparan sulfate exhibits a specific affinity for HMGB1, disrupting the HMGB1/RAGE axis and demonstrating notable hepatoprotective properties.130 The protective effects of berberine-loaded nanostructured lipid carriers and aucubin against HIRI are achieved through the inhibition of the HMGB1/TLR4/NF-κB inflammatory signaling pathway, autophagy, and cell apoptosis.113,198 A newly discovered RG-I pectin-like polysaccharide, YJ3A1, has shown potential in inactivating the HMGB1/TLR4/NF-κB and Akt signaling pathways, impeding the progression of NASH.199 Wang et al. found that the intravenous administration of exosomes derived from adipose-derived mesenchymal stem cells could serve as a safe and effective cellular therapy targeting HMGB1 for HIRI treatment. This approach effectively suppresses the release of HMGB1 by obstructing the TLR4/MyD88/NF-κB/HMGB1 axis, mitigating hepatocyte apoptosis and pyroptosis in a miniature pig model of HIRI.200

HMGB1 is crucial in dampening inflammatory responses, safeguarding liver cells, and impeding the advancement of pathological processes by promoting excessive release and modulating receptor signaling pathways. Although there are challenges to its clinical application, ongoing studies on harnessing HMGB1 as a therapeutic target hold significant promise for effectively treating liver diseases.

Conclusions and future perspectives

Existing studies have confirmed that HMGB1 levels are significantly elevated in the serum of patients with various liver diseases, closely correlating with disease progression, the occurrence of complications, and clinical prognosis. These findings provide a crucial foundation for positioning HMGB1 as a diagnostic and therapeutic target in liver diseases. Furthermore, it is promising that many effective therapeutic drugs targeting HMGB1 in liver disease have been developed. A notable example is traditional Chinese medicine, including curcumin, glycyrrhizin, and salvianic acid A, which exhibit hepatoprotective effects by inhibiting the extracellular release of HMGB1. However, significant challenges remain in translating these findings into clinical interventions. On one hand, most of these clinical discoveries are based on retrospective studies from single centers, making it difficult to extrapolate the conclusions. Large-scale, multicenter prospective studies of HMGB1 in liver diseases are needed for validation. On the other hand, HMGB1 antagonists are still in the preclinical stage. Research on drugs targeting HMGB1 predominantly involves animal subjects, with a lack of studies involving human patient samples. Clinical trials are essential to assess the safety, tolerability, and efficacy of these drugs. Therefore, clinical practice guidelines should be updated, based on the effectiveness and safety of new therapies, to guide physicians in their treatment decisions.

HMGB1 plays a vital role in the pathogenesis of liver diseases; however, its molecular mechanisms of action remain unclear. Understanding the intricate relationship between HMGB1 and disease progression, treatment response, and clinical prognosis is particularly essential. Moreover, the differences in the signaling pathways involving HMGB1 in various hepatic cells, and how these pathways mediate crosstalk between cells, require further exploration. PTMs can modulate HMGB1 functions in liver pathophysiology, and targeting PTMs may offer new therapeutic strategies for the treatment of liver diseases. HMGB1 lactylation represents a highly valuable and challenging research direction. Notably, the specific molecular mechanisms of PTMs—including the writers and erasers, modification sites, and reaction kinetics—remain unclear. The precise signaling pathways and functions of HMGB1 lactylation in regulating the development of liver diseases require further study. Moreover, it remains to be determined whether other liver cells, apart from hepatocytes, can also undergo HMGB1 lactylation. Addressing these challenges will provide new insights into the pathogenesis and treatment of liver diseases.

Declarations

Funding

This study was supported by grants from the Natural Science Foundation of China (82070611), the Guangzhou Science and Technology Plan Projects (2023B03J1287), the Guangdong Medical Science and Technology Research Fund (A2021304), the Five-Year Plan of the Third Affiliated Hospital of Sun Yat-sen University (2024W106), and the Shandong Provincial Natural Science Foundation (ZR2024QH660).

Conflict of interest

The authors have no conflict of interests related to this publication.

Authors’ contributions

Conceptualization, data curation, writing (review & editing) and methodology: LW; conceptualization, writing (original draft preparation), software and formal analysis: ZD; conceptualization, writing (original draft preparation) and literature review: YZ; conceptualization, supervision, visualization, writing (review & editing) and funding acquisition: LP. All authors have read and agreed to the published version of the manuscript.

References

  1. Woolbright BL, Jaeschke H. Role of the inflammasome in acetaminophen-induced liver injury and acute liver failure. J Hepatol 2017;66(4):836-848 View Article PubMed/NCBI
  2. Ramaiah SK, Jaeschke H. Role of neutrophils in the pathogenesis of acute inflammatory liver injury. Toxicol Pathol 2007;35(6):757-766 View Article PubMed/NCBI
  3. Fisher ES, Curry SC. Evaluation and treatment of acetaminophen toxicity. Adv Pharmacol 2019;85:263-272 View Article PubMed/NCBI
  4. Younossi ZM, Stepanova M, Younossi Y, Golabi P, Mishra A, Rafiq N, et al. Epidemiology of chronic liver diseases in the USA in the past three decades. Gut 2020;69(3):564-568 View Article PubMed/NCBI
  5. Julien J, Ayer T, Bethea ED, Tapper EB, Chhatwal J. Projected prevalence and mortality associated with alcohol-related liver disease in the USA, 2019-40: a modelling study. Lancet Public Health 2020;5(6):e316-e323 View Article PubMed/NCBI
  6. Tang D, Kang R, Zeh HJ, Lotze MT. The multifunctional protein HMGB1: 50 years of discovery. Nat Rev Immunol 2023;23(12):824-841 View Article PubMed/NCBI
  7. Alisi A, Nobili V, Ceccarelli S, Panera N, De Stefanis C, De Vito R, et al. Plasma high mobility group box 1 protein reflects fibrosis in pediatric nonalcoholic fatty liver disease. Expert Rev Mol Diagn 2014;14(6):763-771 View Article PubMed/NCBI
  8. Raj R, Shen P, Yu B, Zhang J. A patent review on HMGB1 inhibitors for the treatment of liver diseases. Expert Opin Ther Pat 2024;34(3):127-140 View Article PubMed/NCBI
  9. Xu Z, Xi F, Deng X, Ni Y, Pu C, Wang D, et al. Osteopontin Promotes Macrophage M1 Polarization by Activation of the JAK1/STAT1/HMGB1 Signaling Pathway in Nonalcoholic Fatty Liver Disease. J Clin Transl Hepatol 2023;11(2):273-283 View Article PubMed/NCBI
  10. Saha B, Tornai D, Kodys K, Adejumo A, Lowe P, McClain C, et al. Biomarkers of Macrophage Activation and Immune Danger Signals Predict Clinical Outcomes in Alcoholic Hepatitis. Hepatology 2019;70(4):1134-1149 View Article PubMed/NCBI
  11. Vodovotz Y, Barclay D, Yin J, Squires RH, Zamora R. Dynamics of Systemic Inflammation as a Function of Developmental Stage in Pediatric Acute Liver Failure. Front Immunol 2020;11:610861 View Article PubMed/NCBI
  12. Pil PM, Chow CS, Lippard SJ. High-mobility-group 1 protein mediates DNA bending as determined by ring closures. Proc Natl Acad Sci U S A 1993;90(20):9465-9469 View Article PubMed/NCBI
  13. Bianchi ME, Manfredi AA. High-mobility group box 1 (HMGB1) protein at the crossroads between innate and adaptive immunity. Immunol Rev 2007;220:35-46 View Article PubMed/NCBI
  14. Yang H, Wang H, Chavan SS, Andersson U. High Mobility Group Box Protein 1 (HMGB1): The Prototypical Endogenous Danger Molecule. Mol Med 2015;21(Suppl 1):S6-S12 View Article PubMed/NCBI
  15. Gao B, Wang S, Li J, Han N, Ge H, Zhang G, et al. HMGB1, angel or devil, in ischemic stroke. Brain Behav 2023;13(5):e2987 View Article PubMed/NCBI
  16. Bianchi ME, Falciola L, Ferrari S, Lilley DM. The DNA binding site of HMG1 protein is composed of two similar segments (HMG boxes), both of which have counterparts in other eukaryotic regulatory proteins. EMBO J 1992;11(3):1055-1063 View Article PubMed/NCBI
  17. Starkova TY, Polyanichko AM, Artamonova TO, Tsimokha AS, Tomilin AN, Chikhirzhina EV. Structural Characteristics of High-Mobility Group Proteins HMGB1 and HMGB2 and Their Interaction with DNA. Int J Mol Sci 2023;24(4):3577 View Article PubMed/NCBI
  18. 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(20):5551-5560 View Article PubMed/NCBI
  19. Li J, Kokkola R, Tabibzadeh S, Yang R, Ochani M, Qiang X, et al. Structural basis for the proinflammatory cytokine activity of high mobility group box 1. Mol Med 2003;9(1-2):37-45 View Article PubMed/NCBI
  20. Deng M, Scott MJ, Fan J, Billiar TR. Location is the key to function: HMGB1 in sepsis and trauma-induced inflammation. J Leukoc Biol 2019;106(1):161-169 View Article PubMed/NCBI
  21. Andersson U, Yang H, Harris H. High-mobility group box 1 protein (HMGB1) operates as an alarmin outside as well as inside cells. Semin Immunol 2018;38:40-48 View Article PubMed/NCBI
  22. Chen R, Kang R, Tang D. The mechanism of HMGB1 secretion and release. Exp Mol Med 2022;54(2):91-102 View Article PubMed/NCBI
  23. Tirone M, Tran NL, Ceriotti C, Gorzanelli A, Canepari M, Bottinelli R, et al. High mobility group box 1 orchestrates tissue regeneration via CXCR4. J Exp Med 2018;215(1):303-318 View Article PubMed/NCBI
  24. Haque N, Fareez IM, Fong LF, Mandal C, Abu Kasim NH, Kacharaju KR, et al. Role of the CXCR4-SDF1-HMGB1 pathway in the directional migration of cells and regeneration of affected organs. World J Stem Cells 2020;12(9):938-951 View Article PubMed/NCBI
  25. Tripathi A, Shrinet K, Kumar A. HMGB1 protein as a novel target for cancer. Toxicol Rep 2019;6:253-261 View Article PubMed/NCBI
  26. Kang R, Livesey KM, Zeh HJ, Loze MT, Tang D. HMGB1: a novel Beclin 1-binding protein active in autophagy. Autophagy 2010;6(8):1209-1211 View Article PubMed/NCBI
  27. Ni YA, Chen H, Nie H, Zheng B, Gong Q. HMGB1: An overview of its roles in the pathogenesis of liver disease. J Leukoc Biol 2021;110(5):987-998 View Article PubMed/NCBI
  28. Chen GY, Tang J, Zheng P, Liu Y. CD24 and Siglec-10 selectively repress tissue damage-induced immune responses. Science 2009;323(5922):1722-1725 View Article PubMed/NCBI
  29. Tian J, Avalos AM, Mao SY, Chen B, Senthil K, Wu H, et al. Toll-like receptor 9-dependent activation by DNA-containing immune complexes is mediated by HMGB1 and RAGE. Nat Immunol 2007;8(5):487-496 View Article PubMed/NCBI
  30. Gaskell H, Ge X, Nieto N. High-Mobility Group Box-1 and Liver Disease. Hepatol Commun 2018;2(9):1005-1020 View Article PubMed/NCBI
  31. Nakamoto N, Kanai T. Role of toll-like receptors in immune activation and tolerance in the liver. Front Immunol 2014;5:221 View Article PubMed/NCBI
  32. Tsung A, Sahai R, Tanaka H, Nakao A, Fink MP, Lotze MT, et al. The nuclear factor HMGB1 mediates hepatic injury after murine liver ischemia-reperfusion. J Exp Med 2005;201(7):1135-1143 View Article PubMed/NCBI
  33. Li L, Chen L, Hu L, Liu Y, Sun HY, Tang J, et al. Nuclear factor high-mobility group box1 mediating the activation of Toll-like receptor 4 signaling in hepatocytes in the early stage of nonalcoholic fatty liver disease in mice. Hepatology 2011;54(5):1620-1630 View Article PubMed/NCBI
  34. Cheng LS, Li J, Liu Y, Wang FP, Wang SQ, She WM, et al. HMGB1-induced autophagy: a new pathway to maintain Treg function during chronic hepatitis B virus infection. Clin Sci (Lond) 2017;131(5):381-394 View Article PubMed/NCBI
  35. Li J, Zeng C, Zheng B, Liu C, Tang M, Jiang Y, et al. HMGB1-induced autophagy facilitates hepatic stellate cells activation: a new pathway in liver fibrosis. Clin Sci (Lond) 2018;132(15):1645-1667 View Article PubMed/NCBI
  36. Yamagishi S, Matsui T. Role of receptor for advanced glycation end products (RAGE) in liver disease. Eur J Med Res 2015;20(1):15 View Article PubMed/NCBI
  37. Arriazu E, Ge X, Leung TM, Magdaleno F, Lopategi A, Lu Y, et al. Signalling via the osteopontin and high mobility group box-1 axis drives the fibrogenic response to liver injury. Gut 2017;66(6):1123-1137 View Article PubMed/NCBI
  38. Ge X, Arriazu E, Magdaleno F, Antoine DJ, Dela Cruz R, Theise N, et al. High Mobility Group Box-1 Drives Fibrosis Progression Signaling via the Receptor for Advanced Glycation End Products in Mice. Hepatology 2018;68(6):2380-2404 View Article PubMed/NCBI
  39. Hernandez C, Huebener P, Pradere JP, Antoine DJ, Friedman RA, Schwabe RF. HMGB1 links chronic liver injury to progenitor responses and hepatocarcinogenesis. J Clin Invest 2018;128(6):2436-2451 View Article PubMed/NCBI
  40. Li Y, Qin M, Zhong W, Liu C, Deng G, Yang M, et al. RAGE promotes dysregulation of iron and lipid metabolism in alcoholic liver disease. Redox Biol 2023;59:102559 View Article PubMed/NCBI
  41. Laudenslager M, Lazo M, Wang D, Selvin E, Chen PH, Pankow JS, et al. Association between the soluble receptor for advanced glycation end products (sRAGE) and NAFLD in participants in the Atherosclerosis Risk in Communities Study. Dig Liver Dis 2021;53(7):873-878 View Article PubMed/NCBI
  42. Xie J, Méndez JD, Méndez-Valenzuela V, Aguilar-Hernández MM. Cellular signalling of the receptor for advanced glycation end products (RAGE). Cell Signal 2013;25(11):2185-2197 View Article PubMed/NCBI
  43. Zamaraev AV, Kopeina GS, Prokhorova EA, Zhivotovsky B, Lavrik IN. Post-translational Modification of Caspases: The Other Side of Apoptosis Regulation. Trends Cell Biol 2017;27(5):322-339 View Article PubMed/NCBI
  44. Yu S, Qian L, Ma J. Genetic alterations, RNA expression profiling and DNA methylation of HMGB1 in malignancies. J Cell Mol Med 2022;26(15):4322-4332 View Article PubMed/NCBI
  45. Deribe YL, Pawson T, Dikic I. Post-translational modifications in signal integration. Nat Struct Mol Biol 2010;17(6):666-672 View Article PubMed/NCBI
  46. Shin JH, Kim ID, Kim SW, Lee HK, Jin Y, Park JH, et al. Ethyl pyruvate inhibits HMGB1 phosphorylation and release by chelating calcium. Mol Med 2015;20(1):649-657 View Article PubMed/NCBI
  47. Qi Z, Zhang Y, Qi S, Ling L, Gui L, Yan L, et al. Salidroside Inhibits HMGB1 Acetylation and Release through Upregulation of SirT1 during Inflammation. Oxid Med Cell Longev 2017;2017:9821543 View Article PubMed/NCBI
  48. Ferrara M, Chialli G, Ferreira LM, Ruggieri E, Careccia G, Preti A, et al. Oxidation of HMGB1 Is a Dynamically Regulated Process in Physiological and Pathological Conditions. Front Immunol 2020;11:1122 View Article PubMed/NCBI
  49. Pirnie R, P Gillespie K, Mesaros C, Blair IA. Reappraisal of oxidized HMGB1 as a mediator and biomarker. Future Sci OA 2022;8(10):FSO828 View Article PubMed/NCBI
  50. Kim YH, Kwak MS, Park JB, Lee SA, Choi JE, Cho HS, et al. N-linked glycosylation plays a crucial role in the secretion of HMGB1. J Cell Sci 2016;129(1):29-38 View Article PubMed/NCBI
  51. Yao W, Wang J, Zhu L, Jia X, Xu L, Tian X, et al. Epigenetic Regulator KDM4D Restricts Tumorigenesis via Modulating SYVN1/HMGB1 Ubiquitination Axis in Esophageal Squamous Cell Carcinoma. Front Oncol 2021;11:761346 View Article PubMed/NCBI
  52. 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 View Article PubMed/NCBI
  53. Andersson U, Tracey KJ, Yang H. Post-Translational Modification of HMGB1 Disulfide Bonds in Stimulating and Inhibiting Inflammation. Cells 2021;10(12):3323 View Article PubMed/NCBI
  54. Dong Y, Ming B, Dong L. The Role of HMGB1 in Rheumatic Diseases. Front Immunol 2022;13:815257 View Article PubMed/NCBI
  55. Du S, Zhang X, Jia Y, Peng P, Kong Q, Jiang S, et al. Hepatocyte HSPA12A inhibits macrophage chemotaxis and activation to attenuate liver ischemia/reperfusion injury via suppressing glycolysis-mediated HMGB1 lactylation and secretion of hepatocytes. Theranostics 2023;13(11):3856-3871 View Article PubMed/NCBI
  56. Fu J, Deng W, Ge J, Fu S, Li P, Wu H, et al. Sirtuin 1 alleviates alcoholic liver disease by inhibiting HMGB1 acetylation and translocation. PeerJ 2023;11:e16480 View Article PubMed/NCBI
  57. Tian T, Yao D, Zheng L, Zhou Z, Duan Y, Liu B, et al. Sphingosine kinase 1 regulates HMGB1 translocation by directly interacting with calcium/calmodulin protein kinase II-δ in sepsis-associated liver injury. Cell Death Dis 2020;11(12):1037 View Article PubMed/NCBI
  58. Xue C, Xu Z, Liu Z, Zeng C, Ye Q. Pachymic acid protects hepatic cells against oxygen-glucose deprivation/reperfusion injury by activating sirtuin 1 to inhibit HMGB1 acetylation and inflammatory signaling. Chin J Physiol 2023;66(4):239-247 View Article PubMed/NCBI
  59. Lu B, Wang C, Wang M, Li W, Chen F, Tracey KJ, et al. Molecular mechanism and therapeutic modulation of high mobility group box 1 release and action: an updated review. Expert Rev Clin Immunol 2014;10(6):713-727 View Article PubMed/NCBI
  60. Zhao P, Ye T, Yan X, Hu X, Liu P, Wang X. HMGB1 release by H(2)O(2)-induced hepatocytes is regulated through calcium overload and 58-F interference. Cell Death Discov 2017;3:17008 View Article PubMed/NCBI
  61. Li W, Deng M, Loughran PA, Yang M, Lin M, Yang C, et al. LPS Induces Active HMGB1 Release From Hepatocytes Into Exosomes Through the Coordinated Activities of TLR4 and Caspase-11/GSDMD Signaling. Front Immunol 2020;11:229 View Article PubMed/NCBI
  62. Khanjarsim V, Karimi J, Khodadadi I, Mohammadalipour A, Goodarzi MT, Solgi G, et al. Ameliorative Effects of Nilotinib on CCl4 Induced Liver Fibrosis Via Attenuation of RAGE/HMGB1 Gene Expression and Oxidative Stress in Rat. Chonnam Med J 2017;53(2):118-126 View Article PubMed/NCBI
  63. Xie WH, Ding J, Xie XX, Yang XH, Wu XF, Chen ZX, et al. Hepatitis B virus X protein promotes liver cell pyroptosis under oxidative stress through NLRP3 inflammasome activation. Inflamm Res 2020;69(7):683-696 View Article PubMed/NCBI
  64. Wang X, Xiang L, Li H, Chen P, Feng Y, Zhang J, et al. The Role of HMGB1 Signaling Pathway in the Development and Progression of Hepatocellular Carcinoma: A Review. Int J Mol Sci 2015;16(9):22527-22540 View Article PubMed/NCBI
  65. Tang S, Xu B, Pang H, Xiao L, Mei Q, He X. Ozonated Water Inhibits Hepatocellular Carcinoma Invasion and Metastasis by Regulating the HMGB1/NF-κB/STAT3 Signaling Pathway. J Hepatocell Carcinoma 2023;10:203-215 View Article PubMed/NCBI
  66. Yan W, Chang Y, Liang X, Cardinal JS, Huang H, Thorne SH, et al. High-mobility group box 1 activates caspase-1 and promotes hepatocellular carcinoma invasiveness and metastases. Hepatology 2012;55(6):1863-1875 View Article PubMed/NCBI
  67. Jing M, Xiong X, Mao X, Song Q, Zhang L, Ouyang Y, et al. HMGB1 promotes mitochondrial transfer between hepatocellular carcinoma cells through RHOT1 and RAC1 under hypoxia. Cell Death Dis 2024;15(2):155 View Article PubMed/NCBI
  68. Yang K, Fan M, Wang X, Xu J, Wang Y, Tu F, et al. Lactate promotes macrophage HMGB1 lactylation, acetylation, and exosomal release in polymicrobial sepsis. Cell Death Differ 2022;29(1):133-146 View Article PubMed/NCBI
  69. Kwak MS, Kim HS, Lkhamsuren K, Kim YH, Han MG, Shin JM, et al. Peroxiredoxin-mediated disulfide bond formation is required for nucleocytoplasmic translocation and secretion of HMGB1 in response to inflammatory stimuli. Redox Biol 2019;24:101203 View Article PubMed/NCBI
  70. Habash NW, Sehrawat TS, Shah VH, Cao S. Epigenetics of alcohol-related liver diseases. JHEP Rep 2022;4(5):100466 View Article PubMed/NCBI
  71. Ge X, Antoine DJ, Lu Y, Arriazu E, Leung TM, Klepper AL, et al. High mobility group box-1 (HMGB1) participates in the pathogenesis of alcoholic liver disease (ALD). J Biol Chem 2014;289(33):22672-22691 View Article PubMed/NCBI
  72. Barritt GJ, Litjens TL, Castro J, Aromataris E, Rychkov GY. Store-operated Ca2+ channels and microdomains of Ca2+ in liver cells. Clin Exp Pharmacol Physiol 2009;36(1):77-83 View Article PubMed/NCBI
  73. Arroyave-Ospina JC, Wu Z, Geng Y, Moshage H. Role of Oxidative Stress in the Pathogenesis of Non-Alcoholic Fatty Liver Disease: Implications for Prevention and Therapy. Antioxidants (Basel) 2021;10(2):174 View Article PubMed/NCBI
  74. Zhang D, Tang Z, Huang H, Zhou G, Cui C, Weng Y, et al. Metabolic regulation of gene expression by histone lactylation. Nature 2019;574(7779):575-580 View Article PubMed/NCBI
  75. Gaffney DO, Jennings EQ, Anderson CC, Marentette JO, Shi T, Schou Oxvig AM, et al. Non-enzymatic Lysine Lactoylation of Glycolytic Enzymes. Cell Chem Biol 2020;27(2):206-213.e6 View Article PubMed/NCBI
  76. Brooks GA, Arevalo JA, Osmond AD, Leija RG, Curl CC, Tovar AP. Lactate in contemporary biology: a phoenix risen. J Physiol 2022;600(5):1229-1251 View Article PubMed/NCBI
  77. Liu X, Zhang Y, Li W, Zhou X. Lactylation, an emerging hallmark of metabolic reprogramming: Current progress and open challenges. Front Cell Dev Biol 2022;10:972020 View Article PubMed/NCBI
  78. Pan S, Chen R. Pathological implication of protein post-translational modifications in cancer. Mol Aspects Med 2022;86:101097 View Article PubMed/NCBI
  79. Chu X, Di C, Chang P, Li L, Feng Z, Xiao S, et al. Lactylated Histone H3K18 as a Potential Biomarker for the Diagnosis and Predicting the Severity of Septic Shock. Front Immunol 2021;12:786666 View Article PubMed/NCBI
  80. Gao R, Li Y, Xu Z, Zhang F, Xu J, Hu Y, et al. Mitochondrial pyruvate carrier 1 regulates fatty acid synthase lactylation and mediates treatment of nonalcoholic fatty liver disease. Hepatology 2023;78(6):1800-1815 View Article PubMed/NCBI
  81. Cheng Z, Huang H, Li M, Liang X, Tan Y, Chen Y. Lactylation-Related Gene Signature Effectively Predicts Prognosis and Treatment Responsiveness in Hepatocellular Carcinoma. Pharmaceuticals (Basel) 2023;16(5):644 View Article PubMed/NCBI
  82. Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 2009;324(5930):1029-1033 View Article PubMed/NCBI
  83. Gu J, Zhou J, Chen Q, Xu X, Gao J, Li X, et al. Tumor metabolite lactate promotes tumorigenesis by modulating MOESIN lactylation and enhancing TGF-β signaling in regulatory T cells. Cell Rep 2022;39(12):110986 View Article PubMed/NCBI
  84. Qu J, Li P, Sun Z. Histone lactylation regulates cancer progression by reshaping the tumor microenvironment. Front Immunol 2023;14:1284344 View Article PubMed/NCBI
  85. Pan L, Feng F, Wu J, Fan S, Han J, Wang S, et al. Demethylzeylasteral targets lactate by inhibiting histone lactylation to suppress the tumorigenicity of liver cancer stem cells. Pharmacol Res 2022;181:106270 View Article PubMed/NCBI
  86. Dong V, Nanchal R, Karvellas CJ. Pathophysiology of Acute Liver Failure. Nutr Clin Pract 2020;35(1):24-29 View Article PubMed/NCBI
  87. Lee WM. Acetaminophen (APAP) hepatotoxicity-Isn’t it time for APAP to go away?. J Hepatol 2017;67(6):1324-1331 View Article PubMed/NCBI
  88. Jollow D. Acetaminophen-Induced Hepatic Necrosis: A Reminiscence. Drug Metab Dispos 2024;52(8):707-711 View Article PubMed/NCBI
  89. Zamora R, Barclay D, Yin J, Alonso EM, Leonis MA, Mi Q, et al. HMGB1 is a Central Driver of Dynamic Pro-inflammatory Networks in Pediatric Acute Liver Failure induced by Acetaminophen. Sci Rep 2019;9(1):5971 View Article PubMed/NCBI
  90. Antoine DJ, Williams DP, Kipar A, Jenkins RE, Regan SL, Sathish JG, et al. High-mobility group box-1 protein and keratin-18, circulating serum proteins informative of acetaminophen-induced necrosis and apoptosis in vivo. Toxicol Sci 2009;112(2):521-531 View Article PubMed/NCBI
  91. Pirnie R, Gillespie KP, Weng L, Mesaros C, Blair IA. Characterization and Quantification of Oxidized High Mobility Group Box 1 Proteoforms Secreted from Hepatocytes by Toxic Levels of Acetaminophen. Chem Res Toxicol 2022;35(10):1893-1902 View Article PubMed/NCBI
  92. Yang T, Wang H, Wang X, Li J, Jiang L. The Dual Role of Innate Immune Response in Acetaminophen-Induced Liver Injury. Biology (Basel) 2022;11(7):1057 View Article PubMed/NCBI
  93. Huebener P, Pradere JP, Hernandez C, Gwak GY, Caviglia JM, Mu X, et al. The HMGB1/RAGE axis triggers neutrophil-mediated injury amplification following necrosis. J Clin Invest 2015;125(2):539-550 View Article PubMed/NCBI
  94. Wang X, Sun R, Wei H, Tian Z. High-mobility group box 1 (HMGB1)-Toll-like receptor (TLR)4-interleukin (IL)-23-IL-17A axis in drug-induced damage-associated lethal hepatitis: Interaction of γδ T cells with macrophages. Hepatology 2013;57(1):373-384 View Article PubMed/NCBI
  95. Jiang L, Ke M, Yue S, Xiao W, Yan Y, Deng X, et al. Blockade of Notch signaling promotes acetaminophen-induced liver injury. Immunol Res 2017;65(3):739-749 View Article PubMed/NCBI
  96. Zhang C, Shi X, Su Z, Hu C, Mu X, Pan J, et al. CD36 deficiency ameliorates drug-induced acute liver injury in mice. Mol Med 2021;27(1):57 View Article PubMed/NCBI
  97. Minsart C, Liefferinckx C, Lemmers A, Dressen C, Quertinmont E, Leclercq I, et al. New insights in acetaminophen toxicity: HMGB1 contributes by itself to amplify hepatocyte necrosis in vitro through the TLR4-TRIF-RIPK3 axis. Sci Rep 2020;10(1):5557 View Article PubMed/NCBI
  98. Liu J, Jiang M, Jin Q, Wu YL, Cui ZY, Cui BW, et al. Modulation of HMGB1 Release in APAP-Induced Liver Injury: A Possible Strategy of Chikusetsusaponin V Targeting NETs Formation. Front Pharmacol 2021;12:723881 View Article PubMed/NCBI
  99. Chiew AL, Gluud C, Brok J, Buckley NA. Interventions for paracetamol (acetaminophen) overdose. Cochrane Database Syst Rev 2018;2(2):CD003328 View Article PubMed/NCBI
  100. Lundbäck P, Lea JD, Sowinska A, Ottosson L, Fürst CM, Steen J, et al. A novel high mobility group box 1 neutralizing chimeric antibody attenuates drug-induced liver injury and postinjury inflammation in mice. Hepatology 2016;64(5):1699-1710 View Article PubMed/NCBI
  101. Yang R, Zou X, Tenhunen J, Zhu S, Kajander H, Koskinen ML, et al. HMGB1 neutralization is associated with bacterial translocation during acetaminophen hepatotoxicity. BMC Gastroenterol 2014;14:66 View Article PubMed/NCBI
  102. Du H, Tong S, Kuang G, Gong X, Jiang N, Yang X, et al. Sesamin Protects against APAP-Induced Acute Liver Injury by Inhibiting Oxidative Stress and Inflammatory Response via Deactivation of HMGB1/TLR4/NFκB Signal in Mice. J Immunol Res 2023;2023:1116841 View Article PubMed/NCBI
  103. Jaeschke H. Molecular mechanisms of hepatic ischemia-reperfusion injury and preconditioning. Am J Physiol Gastrointest Liver Physiol 2003;284(1):G15-G26 View Article PubMed/NCBI
  104. Zhai Y, Petrowsky H, Hong JC, Busuttil RW, Kupiec-Weglinski JW. Ischaemia-reperfusion injury in liver transplantation—from bench to bedside. Nat Rev Gastroenterol Hepatol 2013;10(2):79-89 View Article PubMed/NCBI
  105. Mihm S. Danger-Associated Molecular Patterns (DAMPs): Molecular Triggers for Sterile Inflammation in the Liver. Int J Mol Sci 2018;19(10):3104 View Article PubMed/NCBI
  106. 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(33):e2102034118 View Article PubMed/NCBI
  107. Yang M, Antoine DJ, Weemhoff JL, Jenkins RE, Farhood A, Park BK, et al. Biomarkers distinguish apoptotic and necrotic cell death during hepatic ischemia/reperfusion injury in mice. Liver Transpl 2014;20(11):1372-1382 View Article PubMed/NCBI
  108. Feng ZJ, Wang LS, Ma X, Li K, Li XY, Tang Y, et al. Catapol attenuates the aseptic inflammatory response to hepatic I/R injury in vivo and in vitro by inhibiting the HMGB1/TLR-4/NF-κB signaling pathway via the microRNA-410-3p. Mol Immunol 2023;164:66-78 View Article PubMed/NCBI
  109. Tsung A, Klune JR, Zhang X, Jeyabalan G, Cao Z, Peng X, 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 2007;204(12):2913-2923 View Article PubMed/NCBI
  110. Nace GW, Huang H, Klune JR, Eid RE, Rosborough BR, Korff S, et al. Cellular-specific role of toll-like receptor 4 in hepatic ischemia-reperfusion injury in mice. Hepatology 2013;58(1):374-387 View Article PubMed/NCBI
  111. van Golen RF, Reiniers MJ, Marsman G, Alles LK, van Rooyen DM, Petri B, et al. The damage-associated molecular pattern HMGB1 is released early after clinical hepatic ischemia/reperfusion. Biochim Biophys Acta Mol Basis Dis 2019;1865(6):1192-1200 View Article PubMed/NCBI
  112. Koh WU, Kim J, Lee J, Song GW, Hwang GS, Tak E, et al. Remote Ischemic Preconditioning and Diazoxide Protect from Hepatic Ischemic Reperfusion Injury by Inhibiting HMGB1-Induced TLR4/MyD88/NF-κB Signaling. Int J Mol Sci 2019;20(23):5899 View Article PubMed/NCBI
  113. Gendy AM, Elnagar MR, Allam MM, Mousa MR, Khodir AE, El-Haddad AE, et al. Berberine-loaded nanostructured lipid carriers mitigate warm hepatic ischemia/reperfusion-induced lesion through modulation of HMGB1/TLR4/NF-κB signaling and autophagy. Biomed Pharmacother 2022;145:112122 View Article PubMed/NCBI
  114. Xu L, Ge F, Hu Y, Yu Y, Guo K, Miao C. Sevoflurane Postconditioning Attenuates Hepatic Ischemia-Reperfusion Injury by Limiting HMGB1/TLR4/NF-κB Pathway via Modulating microRNA-142 in vivo and in vitro. Front Pharmacol 2021;12:646307 View Article PubMed/NCBI
  115. Du Y, Zhong F, Cheng H, Li T, Chen Y, Tan P, et al. The Dietary Supplement γ-Oryzanol Attenuates Hepatic Ischemia Reperfusion Injury via Inhibiting Endoplasmic Reticulum Stress and HMGB1/NLRP3 Inflammasome. Oxid Med Cell Longev 2021;2021:4628050 View Article PubMed/NCBI
  116. Bai C, Jiang Z, Jiang H, Yu S, Li M, Chu F, et al. Ac2-26 alleviates hepatic ischemia-reperfusion injury based on inhibiting the positive feedback loop of HMGB1/TLR4/NF-κB/neutrophils. Exp Ther Med 2022;24(5):673 View Article PubMed/NCBI
  117. Mcdonald KA, Huang H, Tohme S, Loughran P, Ferrero K, Billiar T, et al. Toll-like receptor 4 (TLR4) antagonist eritoran tetrasodium attenuates liver ischemia and reperfusion injury through inhibition of high-mobility group box protein B1 (HMGB1) signaling. Mol Med 2015;20(1):639-648 View Article PubMed/NCBI
  118. Younis NS, Abdelnaby RM, Mohamed ME. Hepatoprotective effects of linalool against liver ischemia-reperfusion: the role of Nrf2/HO-1/NQO1 and TLR4/RAGE/NFκB pathways. Eur Rev Med Pharmacol Sci 2023;27(20):10094-10111 View Article PubMed/NCBI
  119. Peralta C, Jiménez-Castro MB, Gracia-Sancho J. Hepatic ischemia and reperfusion injury: effects on the liver sinusoidal milieu. J Hepatol 2013;59(5):1094-1106 View Article PubMed/NCBI
  120. Jia K, Zhang Y, Luo R, Liu R, Li Y, Wu J, et al. Acteoside ameliorates hepatic ischemia-reperfusion injury via reversing the senescent fate of liver sinusoidal endothelial cells and restoring compromised sinusoidal networks. Int J Biol Sci 2023;19(15):4967-4988 View Article PubMed/NCBI
  121. Noguchi D, Kuriyama N, Hibi T, Maeda K, Shinkai T, Gyoten K, et al. The Impact of Dabigatran Treatment on Sinusoidal Protection Against Hepatic Ischemia/Reperfusion Injury in Mice. Liver Transpl 2021;27(3):363-384 View Article PubMed/NCBI
  122. Vasques F, Cavazza A, Bernal W. Acute liver failure. Curr Opin Crit Care 2022;28(2):198-207 View Article PubMed/NCBI
  123. Yang R, Zou X, Tenhunen J, Tønnessen TI. HMGB1 and Extracellular Histones Significantly Contribute to Systemic Inflammation and Multiple Organ Failure in Acute Liver Failure. Mediators Inflamm 2017;2017:5928078 View Article PubMed/NCBI
  124. Luo L, Wang S, Chen B, Zhong M, Du R, Wei C, et al. Inhibition of inflammatory liver injury by the HMGB1-A box through HMGB1/TLR-4/NF-κB signaling in an acute liver failure mouse model. Front Pharmacol 2022;13:990087 View Article PubMed/NCBI
  125. Liu Y, Yuan W, Fang M, Guo H, Zhang X, Mei X, et al. Determination of HMGB1 in hepatitis B virus-related acute-on-chronic liver failure patients with acute kidney injury: Early prediction and prognostic implications. Front Pharmacol 2022;13:1031790 View Article PubMed/NCBI
  126. Duan M, Liu X, Yang Y, Zhang Y, Wu R, Lv Y, et al. Orchestrated regulation of immune inflammation with cell therapy in pediatric acute liver injury. Front Immunol 2023;14:1194588 View Article PubMed/NCBI
  127. Wang Y, Zhang H, Chen Q, Jiao F, Shi C, Pei M, et al. TNF-α/HMGB1 inflammation signalling pathway regulates pyroptosis during liver failure and acute kidney injury. Cell Prolif 2020;53(6):e12829 View Article PubMed/NCBI
  128. Wang G, Jin S, Huang W, Li Y, Wang J, Ling X, et al. LPS-induced macrophage HMGB1-loaded extracellular vesicles trigger hepatocyte pyroptosis by activating the NLRP3 inflammasome. Cell Death Discov 2021;7(1):337 View Article PubMed/NCBI
  129. Wang Y, Chen Q, Shi C, Jiao F, Gong Z. Mechanism of glycyrrhizin on ferroptosis during acute liver failure by inhibiting oxidative stress. Mol Med Rep 2019;20(5):4081-4090 View Article PubMed/NCBI
  130. Arnold K, Xu Y, Sparkenbaugh EM, Li M, Han X, Zhang X, et al. Design of anti-inflammatory heparan sulfate to protect against acetaminophen-induced acute liver failure. Sci Transl Med 2020;12(535):eaav8075 View Article PubMed/NCBI
  131. Ye Q, Zou B, Yeo YH, Li J, Huang DQ, Wu Y, et al. Global prevalence, incidence, and outcomes of non-obese or lean non-alcoholic fatty liver disease: a systematic review and meta-analysis. Lancet Gastroenterol Hepatol 2020;5(8):739-752 View Article PubMed/NCBI
  132. Younossi Z, Anstee QM, Marietti M, Hardy T, Henry L, Eslam M, et al. Global burden of NAFLD and NASH: trends, predictions, risk factors and prevention. Nat Rev Gastroenterol Hepatol 2018;15(1):11-20 View Article PubMed/NCBI
  133. Yates KP, Deppe R, Comerford M, Masuoka H, Cummings OW, Tonascia J, et al. Serum high mobility group box 1 protein levels are not associated with either histological severity or treatment response in children and adults with nonalcoholic fatty liver disease. PLoS One 2017;12(11):e0185813 View Article PubMed/NCBI
  134. Puchades Renau L, Herreras López J, Cebrià I, Iranzo MÀ, Cezón Serrano N, Di Maira T, Berenguer M. Frailty and Sarcopenia in Acute-on-Chronic Liver Failure. Hepatol Commun 2021;5(8):1333-1347 View Article PubMed/NCBI
  135. Tandon P, Montano-Loza AJ, Lai JC, Dasarathy S, Merli M. Sarcopenia and frailty in decompensated cirrhosis. J Hepatol 2021;75(Suppl 1):S147-S162 View Article PubMed/NCBI
  136. Lee YH, Kim SU, Song K, Park JY, Kim DY, Ahn SH, et al. Sarcopenia is associated with significant liver fibrosis independently of obesity and insulin resistance in nonalcoholic fatty liver disease: Nationwide surveys (KNHANES 2008-2011). Hepatology 2016;63(3):776-786 View Article PubMed/NCBI
  137. Ohmori H, Kawahara I, Mori T, Nukaga S, Luo Y, Kishi S, et al. Evaluation of Parameters for Cancer-Induced Sarcopenia in Patients Autopsied after Death from Colorectal Cancer. Pathobiology 2019;86(5-6):306-314 View Article PubMed/NCBI
  138. Narasimhulu CA, Singla DK. BMP-7 Attenuates Sarcopenia and Adverse Muscle Remodeling in Diabetic Mice via Alleviation of Lipids, Inflammation, HMGB1, and Pyroptosis. Antioxidants (Basel) 2023;12(2):331 View Article PubMed/NCBI
  139. Ho TL, Tang CH, Chang SL, Tsai CH, Chen HT, Su CM. HMGB1 Promotes In Vitro and In Vivo Skeletal Muscle Atrophy through an IL-18-Dependent Mechanism. Cells 2022;11(23):3936 View Article PubMed/NCBI
  140. Chen X, Ling Y, Wei Y, Tang J, Ren Y, Zhang B, et al. Dual regulation of HMGB1 by combined JNK1/2-ATF2 axis with miR-200 family in nonalcoholic steatohepatitis in mice. FASEB J 2018;32(5):2722-2734 View Article PubMed/NCBI
  141. Zhang C, Chen F, Feng L, Shan Q, Zheng GH, Wang YJ, et al. FBXW7 suppresses HMGB1-mediated innate immune signaling to attenuate hepatic inflammation and insulin resistance in a mouse model of nonalcoholic fatty liver disease. Mol Med 2019;25(1):29 View Article PubMed/NCBI
  142. Zeng W, Shan W, Gao L, Gao D, Hu Y, Wang G, et al. Inhibition of HMGB1 release via salvianolic acid B-mediated SIRT1 up-regulation protects rats against non-alcoholic fatty liver disease. Sci Rep 2015;5:16013 View Article PubMed/NCBI
  143. Lin M, Long J, Li W, Yang C, Loughran P, O’Doherty R, et al. Hepatocyte high-mobility group box 1 protects against steatosis and cellular stress during high fat diet feeding. Mol Med 2020;26(1):115 View Article PubMed/NCBI
  144. Personnaz J, Piccolo E, Dortignac A, Iacovoni JS, Mariette J, Rocher V, et al. Nuclear HMGB1 protects from nonalcoholic fatty liver disease through negative regulation of liver X receptor. Sci Adv 2022;8(12):eabg9055 View Article PubMed/NCBI
  145. Zhang X, Lin Y, Lin S, Li C, Gao J, Feng Z, et al. Silencing of functional p53 attenuates NAFLD by promoting HMGB1-related autophagy induction. Hepatol Int 2020;14(5):828-841 View Article PubMed/NCBI
  146. Dukić M, Radonjić T, Jovanović I, Zdravković M, Todorović Z, Kraišnik N, et al. Alcohol, Inflammation, and Microbiota in Alcoholic Liver Disease. Int J Mol Sci 2023;24(4):3735 View Article PubMed/NCBI
  147. Bukong TN, Cho Y, Iracheta-Vellve A, Saha B, Lowe P, Adejumo A, et al. Abnormal neutrophil traps and impaired efferocytosis contribute to liver injury and sepsis severity after binge alcohol use. J Hepatol 2018;69(5):1145-1154 View Article PubMed/NCBI
  148. Gan LT, Van Rooyen DM, Koina ME, McCuskey RS, Teoh NC, Farrell GC. Hepatocyte free cholesterol lipotoxicity results from JNK1-mediated mitochondrial injury and is HMGB1 and TLR4-dependent. J Hepatol 2014;61(6):1376-1384 View Article PubMed/NCBI
  149. Seo YS, Kwon JH, Yaqoob U, Yang L, De Assuncao TM, Simonetto DA, et al. HMGB1 recruits hepatic stellate cells and liver endothelial cells to sites of ethanol-induced parenchymal cell injury. Am J Physiol Gastrointest Liver Physiol 2013;305(11):G838-G848 View Article PubMed/NCBI
  150. Xie ZY, Xiao ZH, Wang FF. Inhibition of autophagy reverses alcohol-induced hepatic stellate cells activation through activation of Nrf2-Keap1-ARE signaling pathway. Biochimie 2018;147:55-62 View Article PubMed/NCBI
  151. Xie ZY, Wang FF, Xiao ZH, Liu SF, Lai YL, Tang SL. Long noncoding RNA XIST enhances ethanol-induced hepatic stellate cells autophagy and activation via miR-29b/HMGB1 axis. IUBMB Life 2019;71(12):1962-1972 View Article PubMed/NCBI
  152. Lan Y, Yan R, Shan W, Chu J, Sun R, Wang R, et al. Salvianic acid A alleviates chronic alcoholic liver disease by inhibiting HMGB1 translocation via down-regulating BRD4. J Cell Mol Med 2020;24(15):8518-8531 View Article PubMed/NCBI
  153. Shang Y, Jiang M, Chen N, Jiang XL, Zhan ZY, Zhang ZH, et al. Inhibition of HMGB1/TLR4 Signaling Pathway by Digitoflavone: A Potential Therapeutic Role in Alcohol-Associated Liver Disease. J Agric Food Chem 2022;70(9):2968-2983 View Article PubMed/NCBI
  154. Kisseleva T, Brenner D. Molecular and cellular mechanisms of liver fibrosis and its regression. Nat Rev Gastroenterol Hepatol 2021;18(3):151-166 View Article PubMed/NCBI
  155. He Q, Fu Y, Ding X, Li D, Wang Z, Tian D, et al. High-mobility group box 1 induces endoplasmic reticulum stress and activates hepatic stellate cells. Lab Invest 2018;98(9):1200-1210 View Article PubMed/NCBI
  156. Zhong H, Gui X, Hou L, Lv R, Jin Y. From Inflammation to Fibrosis: Novel Insights into the Roles of High Mobility Group Protein Box 1 in Schistosome-Induced Liver Damage. Pathogens 2022;11(3):289 View Article PubMed/NCBI
  157. de Oliveira Gomes CG, de Andrade MVM, Guedes LR, Rocha HC, Guimarães RG, Carvalho FAC, et al. Evaluation of the Biomarkers HMGB1 and IL-6 as Predictors of Mortality in Cirrhotic Patients with Acute Kidney Injury. Mediators Inflamm 2020;2020:2867241 View Article PubMed/NCBI
  158. Ho WI, Hu Y, Cheng CW, Wei R, Yang J, Li N, et al. Liposome-encapsulated curcumin attenuates HMGB1-mediated hepatic inflammation and fibrosis in a murine model of Wilson’s disease. Biomed Pharmacother 2022;152:113197 View Article PubMed/NCBI
  159. Abdelfattah AM, Mahmoud SS, El-Wafaey DI, Abdelgeleel HM, Abdelhamid AM. Diacerein ameliorates cholestasis-induced liver fibrosis in rat via modulating HMGB1/RAGE/NF-κB/JNK pathway and endoplasmic reticulum stress. Sci Rep 2023;13(1):11455 View Article PubMed/NCBI
  160. Zeng HH, Ma M, Wang YL, Chen MH, Huang DB. Hyperoside attenuates carbon tetrachloride-induced hepatic fibrosis via the poly(ADP-ribose)polymerase-1-high mobility group protein 1 pathway. Eur J Pharmacol 2023;960:176178 View Article PubMed/NCBI
  161. Zhao T, Zhu Y, Yao L, Liu L, Li N. IGF-1 alleviates CCL4-induced hepatic cirrhosis and dysfunction of intestinal barrier through inhibition TLR4/NF-κB signaling mediated by down-regulation HMGB1. Ann Hepatol 2021;26:100560 View Article PubMed/NCBI
  162. Yang H, Ochani M, Li J, Qiang X, Tanovic M, Harris HE, et al. Reversing established sepsis with antagonists of endogenous high-mobility group box 1. Proc Natl Acad Sci U S A 2004;101(1):296-301 View Article PubMed/NCBI
  163. Palumbo R, Sampaolesi M, De Marchis F, Tonlorenzi R, Colombetti S, Mondino A, et al. Extracellular HMGB1, a signal of tissue damage, induces mesoangioblast migration and proliferation. J Cell Biol 2004;164(3):441-449 View Article PubMed/NCBI
  164. Nojiri S, Tsuchiya A, Natsui K, Takeuchi S, Watanabe T, Kojima Y, et al. Synthesized HMGB1 peptide attenuates liver inflammation and suppresses fibrosis in mice. Inflamm Regen 2021;41(1):28 View Article PubMed/NCBI
  165. He M, Chu T, Wang Z, Feng Y, Shi R, He M, et al. Inhibition of macrophages inflammasome activation via autophagic degradation of HMGB1 by EGCG ameliorates HBV-induced liver injury and fibrosis. Front Immunol 2023;14:1147379 View Article PubMed/NCBI
  166. Huang Y, Liangpunsakul S, Rudraiah S, Ma J, Keshipeddy SK, Wright D, et al. HMGB2 is a potential diagnostic marker and therapeutic target for liver fibrosis and cirrhosis. Hepatol Commun 2023;7(11):e0299 View Article PubMed/NCBI
  167. Ringelhan M, Heikenwalder M, Protzer U. Direct effects of hepatitis B virus-encoded proteins and chronic infection in liver cancer development. Dig Dis 2013;31(1):138-151 View Article PubMed/NCBI
  168. Cheng BQ, Jia CQ, Liu CT, Lu XF, Zhong N, Zhang ZL, et al. Serum high mobility group box chromosomal protein 1 is associated with clinicopathologic features in patients with hepatocellular carcinoma. Dig Liver Dis 2008;40(6):446-452 View Article PubMed/NCBI
  169. Dong YD, Cui L, Peng CH, Cheng DF, Han BS, Huang F. Expression and clinical significance of HMGB1 in human liver cancer: Knockdown inhibits tumor growth and metastasis in vitro and in vivo. Oncol Rep 2013;29(1):87-94 View Article PubMed/NCBI
  170. Khambu B, Huda N, Chen X, Antoine DJ, Li Y, Dai G, et al. HMGB1 promotes ductular reaction and tumorigenesis in autophagy-deficient livers. J Clin Invest 2018;128(6):2419-2435 View Article PubMed/NCBI
  171. Athavale D, Song Z, Desert R, Han H, Das S, Ge X, et al. Ablation of high-mobility group box-1 in the liver reduces hepatocellular carcinoma but causes hyperbilirubinemia in Hippo signaling-deficient mice. Hepatol Commun 2022;6(8):2155-2169 View Article PubMed/NCBI
  172. Wei Y, Tang X, Ren Y, Yang Y, Song F, Fu J, et al. An RNA-RNA crosstalk network involving HMGB1 and RICTOR facilitates hepatocellular carcinoma tumorigenesis by promoting glutamine metabolism and impedes immunotherapy by PD-L1+ exosomes activity. Signal Transduct Target Ther 2021;6(1):421 View Article PubMed/NCBI
  173. Chen Y, Lin C, Liu Y, Jiang Y. HMGB1 promotes HCC progression partly by downregulating p21 via ERK/c-Myc pathway and upregulating MMP-2. Tumour Biol 2016;37(4):4399-4408 View Article PubMed/NCBI
  174. Yang Y, Zhao LH, Huang B, Wang RY, Yuan SX, Tao QF, et al. Pioglitazone, a PPARγ agonist, inhibits growth and invasion of human hepatocellular carcinoma via blockade of the rage signaling. Mol Carcinog 2015;54(12):1584-1595 View Article PubMed/NCBI
  175. Cheng P, Dai W, Wang F, Lu J, Shen M, Chen K, et al. Ethyl pyruvate inhibits proliferation and induces apoptosis of hepatocellular carcinoma via regulation of the HMGB1-RAGE and AKT pathways. Biochem Biophys Res Commun 2014;443(4):1162-1168 View Article PubMed/NCBI
  176. Fan A, Gao M, Tang X, Jiao M, Wang C, Wei Y, et al. HMGB1/RAGE axis in tumor development: unraveling its significance. Front Oncol 2024;14:1336191 View Article PubMed/NCBI
  177. Feng W, Chen J, Huang W, Wang G, Chen X, Duan L, et al. HMGB1-mediated elevation of KLF7 facilitates hepatocellular carcinoma progression and metastasis through upregulating TLR4 and PTK2. Theranostics 2023;13(12):4042-4058 View Article PubMed/NCBI
  178. Khambu B, Hong H, Liu S, Liu G, Chen X, Dong Z, et al. The HMGB1-RAGE axis modulates the growth of autophagy-deficient hepatic tumors. Cell Death Dis 2020;11(5):333 View Article PubMed/NCBI
  179. Pu Z, Duda DG, Zhu Y, Pei S, Wang X, Huang Y, et al. VCP interaction with HMGB1 promotes hepatocellular carcinoma progression by activating the PI3K/AKT/mTOR pathway. J Transl Med 2022;20(1):212 View Article PubMed/NCBI
  180. Jin S, Yang Z, Hao X, Tang W, Ma W, Zong H. Roles of HMGB1 in regulating myeloid-derived suppressor cells in the tumor microenvironment. Biomark Res 2020;8:21 View Article PubMed/NCBI
  181. Jiang J, Wang GZ, Wang Y, Huang HZ, Li WT, Qu XD. Hypoxia-induced HMGB1 expression of HCC promotes tumor invasiveness and metastasis via regulating macrophage-derived IL-6. Exp Cell Res 2018;367(1):81-88 View Article PubMed/NCBI
  182. Zhang Y, Ren H, Li J, Xue R, Liu H, Zhu Z, et al. Elevated HMGB1 expression induced by hepatitis B virus X protein promotes epithelial-mesenchymal transition and angiogenesis through STAT3/miR-34a/NF-κB in primary liver cancer. Am J Cancer Res 2021;11(2):479-494 View Article PubMed/NCBI
  183. Takino J, Yamagishi S, Takeuchi M. Glycer-AGEs-RAGE signaling enhances the angiogenic potential of hepatocellular carcinoma by upregulating VEGF expression. World J Gastroenterol 2012;18(15):1781-1788 View Article PubMed/NCBI
  184. Xiao Y, Sun L, Fu Y, Huang Y, Zhou R, Hu X, et al. High mobility group box 1 promotes sorafenib resistance in HepG2 cells and in vivo. BMC Cancer 2017;17(1):857 View Article PubMed/NCBI
  185. Hubert P, Roncarati P, Demoulin S, Pilard C, Ancion M, Reynders C, et al. Extracellular HMGB1 blockade inhibits tumor growth through profoundly remodeling immune microenvironment and enhances checkpoint inhibitor-based immunotherapy. J Immunother Cancer 2021;9(3):e001966 View Article PubMed/NCBI
  186. Wang ZH, Chen L, Li W, Chen L, Wang YP. Mitochondria transfer and transplantation in human health and diseases. Mitochondrion 2022;65:80-87 View Article PubMed/NCBI
  187. Xue J, Suarez JS, Minaai M, Li S, Gaudino G, Pass HI, et al. HMGB1 as a therapeutic target in disease. J Cell Physiol 2021;236(5):3406-3419 View Article PubMed/NCBI
  188. Andersson U, Yang H, Harris H. Extracellular HMGB1 as a therapeutic target in inflammatory diseases. Expert Opin Ther Targets 2018;22(3):263-277 View Article PubMed/NCBI
  189. Yang H, Wang H, Andersson U. Targeting Inflammation Driven by HMGB1. Front Immunol 2020;11:484 View Article PubMed/NCBI
  190. 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(1):13 View Article PubMed/NCBI
  191. Hua S, Ma M, Fei X, Zhang Y, Gong F, Fang M. Glycyrrhizin attenuates hepatic ischemia-reperfusion injury by suppressing HMGB1-dependent GSDMD-mediated kupffer cells pyroptosis. Int Immunopharmacol 2019;68:145-155 View Article PubMed/NCBI
  192. Yang H, Wang H, Ju Z, Ragab AA, Lundbäck P, Long W, et al. MD-2 is required for disulfide HMGB1-dependent TLR4 signaling. J Exp Med 2015;212(1):5-14 View Article PubMed/NCBI
  193. Lv G, Yang M, Gai K, Jia Q, Wang Z, Wang B, et al. Multiple functions of HMGB1 in cancer. Front Oncol 2024;14:1384109 View Article PubMed/NCBI
  194. Lu L, Qiu C, Li D, Bai G, Liang J, Yang Q. MicroRNA-505 suppresses proliferation and invasion in hepatoma cells by directly targeting high-mobility group box 1. Life Sci 2016;157:12-18 View Article PubMed/NCBI
  195. Lv G, Wu M, Wang M, Jiang X, Du J, Zhang K, et al. miR-320a regulates high mobility group box 1 expression and inhibits invasion and metastasis in hepatocellular carcinoma. Liver Int 2017;37(9):1354-1364 View Article PubMed/NCBI
  196. Liu Z, Dou C, Yao B, Xu M, Ding L, Wang Y, et al. Methylation-mediated repression of microRNA-129-2 suppresses cell aggressiveness by inhibiting high mobility group box 1 in human hepatocellular carcinoma. Oncotarget 2016;7(24):36909-36923 View Article PubMed/NCBI
  197. Zhang DY, Zou XJ, Cao CH, Zhang T, Lei L, Qi XL, et al. Identification and Functional Characterization of Long Non-coding RNA MIR22HG as a Tumor Suppressor for Hepatocellular Carcinoma. Theranostics 2018;8(14):3751-3765 View Article PubMed/NCBI
  198. Zhang S, Feng Z, Gao W, Duan Y, Fan G, Geng X, et al. Aucubin Attenuates Liver Ischemia-Reperfusion Injury by Inhibiting the HMGB1/TLR-4/NF-κB Signaling Pathway, Oxidative Stress, and Apoptosis. Front Pharmacol 2020;11:544124 View Article PubMed/NCBI
  199. Jing X, Zhou G, Zhu A, Jin C, Li M, Ding K. RG-I pectin-like polysaccharide from Rosa chinensis inhibits inflammation and fibrosis associated to HMGB1/TLR4/NF-κB signaling pathway to improve non-alcoholic steatohepatitis. Carbohydr Polym 2024;337:122139 View Article PubMed/NCBI
  200. Wang Y, Piao C, Liu T, Lu X, Ma Y, Zhang J, et al. Effects of the exosomes of adipose-derived mesenchymal stem cells on apoptosis and pyroptosis of injured liver in miniature pigs. Biomed Pharmacother 2023;169:115873 View Article PubMed/NCBI