Display options
Share it on

Acta Pharmacol Sin. 2021 May 26; doi: 10.1038/s41401-021-00676-7. Epub 2021 May 26.

Targeting HMGB1 for the treatment of sepsis and sepsis-induced organ injury.

Acta pharmacologica Sinica

Chao Deng, Lin Zhao, Zhi Yang, Jia-Jia Shang, Chang-Yu Wang, Ming-Zhi Shen, Shuai Jiang, Tian Li, Wen-Cheng Di, Ying Chen, He Li, Ye-Dong Cheng, Yang Yang

Affiliations

  1. Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China.
  2. Department of Orthopaedics, Huaian Medical District of Jingling Hospital, Medical School of Nanjing University, Huaian, 213001, China.
  3. Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
  4. Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China.
  5. Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Life of Sciences, Northwest University, Xi'an, 710021, China.
  6. Department of Cardiology, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China.
  7. Hainan Hospital of PLA General Hospital, The Second School of Clinical Medicine, Southern Medical University, Sanya, 572013, China.
  8. National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, 518100, China.
  9. Department of Hematology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
  10. Department of Orthopaedics, Huaian Medical District of Jingling Hospital, Medical School of Nanjing University, Huaian, 213001, China. [email protected].
  11. Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, Xi'an, 710021, China. [email protected].
  12. Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Life of Sciences, Northwest University, Xi'an, 710021, China. [email protected].

PMID: 34040166 DOI: 10.1038/s41401-021-00676-7

Abstract

High mobility group box 1 (HMGB1) is a ubiquitous nuclear protein that is present in almost all cells and regulates the activity of innate immune responses in both intracellular and extracellular settings. Current evidence suggests that HMGB1 plays a pivotal role in human pathological and pathophysiological processes such as the inflammatory response, immune reactions, cell migration, aging, and cell death. Sepsis is a systemic inflammatory response syndrome (SIRS) that occurs in hosts in response to microbial infections with a proven or suspected infectious etiology and is the leading cause of death in intensive care units worldwide, particularly in the aging population. Dysregulated systemic inflammation is a classic characteristic of sepsis, and suppression of HMGB1 may ameliorate inflammation and improve patient outcomes. Here, we focus on the latest breakthroughs regarding the roles of HMGB1 in sepsis and sepsis-related organ injury, the ways by which HMGB1 are released, and the signaling pathways and therapeutics associated with HMGB1. This review highlights recent advances related to HMGB1: the regulation of HMBG1 might be helpful for both basic research and drug development for the treatment of sepsis and sepsis-related organ injury.

Keywords: LPS; Toll-like receptor; high mobility group box 1; inflammation; sepsis

References

  1. Sims GP, Rowe DC, Rietdijk ST, Herbst R, Coyle AJ. HMGB1 and RAGE in inflammation and cancer. Annu Rev Immunol. 2010;28:367–88. - PubMed
  2. Hu W, Deng C, Ma Z, Wang D, Fan C, Li T, et al. Utilizing melatonin to combat bacterial infections and septic injury. Br J Pharmacol. 2017;174:754–68. - PubMed
  3. Hardeland R. Melatonin and inflammation-story of a double-edged blade. J Pineal Res. 2018;65:e12525. - PubMed
  4. Xu D, Liao S, Li P, Zhang Q, Lv Y, Fu X, et al. Metabolomics coupled with transcriptomics approach deciphering age relevance in sepsis. Aging Dis. 2019;10:854–70. - PubMed
  5. da Rocha EP, Yokota LG, Sampaio BM, Cardoso Eid KZ, Dias DB, de Freitas FM, et al. Urinary neutrophil gelatinase-associated lipocalin is excellent predictor of acute kidney injury in septic elderly patients. Aging Dis. 2018;9:182–91. - PubMed
  6. Gotts JE, Matthay MA. Sepsis: pathophysiology and clinical management. BMJ. 2016;353:i1585. - PubMed
  7. Tracey KJ, Beutler B, Lowry SF, Merryweather J, Wolpe S, Milsark IW, et al. Shock and tissue injury induced by recombinant human cachectin. Science. 1986;234:470–4. - PubMed
  8. Wang H, Bloom O, Zhang M, Vishnubhakat JM, Ombrellino M, Che J, et al. HMG-1 as a late mediator of endotoxin lethality in mice. Science. 1999;285:248–51. - PubMed
  9. Wang H, Ward MF, Sama AE. Targeting HMGB1 in the treatment of sepsis. Expert Opin Ther Targets. 2014;18:257–68. - PubMed
  10. Zeni F, Freeman B, Natanson C. Anti-inflammatory therapies to treat sepsis and septic shock: a reassessment. Crit Care Med. 1997;25:1095–100. - PubMed
  11. Einck L, Bustin M. The intracellular distribution and function of the high mobility group chromosomal proteins. Exp Cell Res. 1985;156:295–310. - PubMed
  12. Javaherian K, Liu JF, Wang JC. Nonhistone proteins HMG1 and HMG2 change the DNA helical structure. Science. 1978;199:1345–6. - PubMed
  13. Yang H, Antoine DJ, Andersson U, Tracey KJ. The many faces of HMGB1: molecular structure-functional activity in inflammation, apoptosis, and chemotaxis. J Leukoc Biol. 2013;93:865–73. - PubMed
  14. Andersson U, Tracey KJ. HMGB1 in sepsis. Scand J Infect Dis. 2003;35:577–84. - PubMed
  15. Gregoire M, Tadie JM, Uhel F, Gacouin A, Piau C, Bone N, et al. Frontline science: HMGB1 induces neutrophil dysfunction in experimental sepsis and in patients who survive septic shock. J Leukoc Biol. 2017;101:1281–7. - PubMed
  16. Hotchkiss RS, Karl IE. The pathophysiology and treatment of sepsis. N Engl J Med. 2003;348:138–50. - PubMed
  17. Efron PA, Martins A, Minnich D, Tinsley K, Ungaro R, Bahjat FR, et al. Characterization of the systemic loss of dendritic cells in murine lymph nodes during polymicrobial sepsis. J Immunol. 2004;173:3035–43. - PubMed
  18. Cheng SC, Scicluna BP, Arts RJ, Gresnigt MS, Lachmandas E, Giamarellos-Bourboulis EJ, et al. Broad defects in the energy metabolism of leukocytes underlie immunoparalysis in sepsis. Nat Immunol. 2016;17:406–13. - PubMed
  19. Hotchkiss RS, Monneret G, Payen D. Sepsis-induced immunosuppression: from cellular dysfunctions to immunotherapy. Nat Rev Immunol. 2013;13:862–74. - PubMed
  20. Buwalda M, Ince C. Opening the microcirculation: can vasodilators be useful in sepsis? Intensive Care Med. 2002;28:1208–17. - PubMed
  21. Escames G, Lopez LC, Ortiz F, Lopez A, Garcia JA, Ros E, et al. Attenuation of cardiac mitochondrial dysfunction by melatonin in septic mice. FEBS J. 2007;274:2135–47. - PubMed
  22. Singer M. The role of mitochondrial dysfunction in sepsis-induced multi-organ failure. Virulence. 2014;5:66–72. - PubMed
  23. Aronis A, Aharoni-Simon M, Madar Z, Tirosh O. Triacylglycerol-induced impairment in mitochondrial biogenesis and function in J774.2 and mouse peritoneal macrophage foam cells. Arch Biochem Biophys. 2009;492:74–81. - PubMed
  24. Wu Y, Yao YM, Lu ZQ. Mitochondrial quality control mechanisms as potential therapeutic targets in sepsis-induced multiple organ failure. J Mol Med (Berl). 2019;97:451–62. - PubMed
  25. Cecconi M, Evans L, Levy M, Rhodes A. Sepsis and septic shock. Lancet. 2018;392:75–87. - PubMed
  26. Biemond BJ, Levi M, Ten Cate H, Van der Poll T, Buller HR, Hack CE, et al. Plasminogen activator and plasminogen activator inhibitor I release during experimental endotoxaemia in chimpanzees: effect of interventions in the cytokine and coagulation cascades. Clin Sci (Lond). 1995;88:587–94. - PubMed
  27. Muscatell KA, Dedovic K, Slavich GM, Jarcho MR, Breen EC, Bower JE, et al. Greater amygdala activity and dorsomedial prefrontal-amygdala coupling are associated with enhanced inflammatory responses to stress. Brain Behav Immun. 2015;43:46–53. - PubMed
  28. Sonneville R, Verdonk F, Rauturier C, Klein IF, Wolff M, Annane D, et al. Understanding brain dysfunction in sepsis. Ann Intensive Care. 2013;3:15. - PubMed
  29. Kanczkowski W, Sue M, Zacharowski K, Reincke M, Bornstein SR. The role of adrenal gland microenvironment in the HPA axis function and dysfunction during sepsis. Mol Cell Endocrinol. 2015;408:241–8. - PubMed
  30. Tynan RJ, Weidenhofer J, Hinwood M, Cairns MJ, Day TA, Walker FR. A comparative examination of the anti-inflammatory effects of SSRI and SNRI antidepressants on LPS stimulated microglia. Brain Behav Immun. 2012;26:469–79. - PubMed
  31. Fujii T, Mashimo M, Moriwaki Y, Misawa H, Ono S, Horiguchi K, et al. Expression and function of the cholinergic system in immune cells. Front Immunol. 2017;8:1085. - PubMed
  32. Oakes SA, Papa FR. The role of endoplasmic reticulum stress in human pathology. Annu Rev Pathol. 2015;10:173–94. - PubMed
  33. Li Y, Guo Y, Tang J, Jiang J, Chen Z. New insights into the roles of CHOP-induced apoptosis in ER stress. Acta Biochim Biophys Sin (Shanghai). 2014;46:629–40. - PubMed
  34. Garcia de la Cadena S, Massieu L. Caspases and their role in inflammation and ischemic neuronal death. Focus on caspase-12. Apoptosis. 2016;21:763–77. - PubMed
  35. Jiao G, Hao L, Wang M, Zhong B, Yu M, Zhao S, et al. Upregulation of endoplasmic reticulum stress is associated with diaphragm contractile dysfunction in a rat model of sepsis. Mol Med Rep. 2017;15:366–74. - PubMed
  36. Ho J, Yu J, Wong SH, Zhang L, Liu X, Wong WT, et al. Autophagy in sepsis: degradation into exhaustion? Autophagy. 2016;12:1073–82. - PubMed
  37. Schafer ST, Franken L, Adamzik M, Schumak B, Scherag A, Engler A, et al. Mitochondrial DNA: an endogenous trigger for immune paralysis. Anesthesiology. 2016;124:923–33. - PubMed
  38. Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell. 2010;140:805–20. - PubMed
  39. Schulte W, Bernhagen J, Bucala R. Cytokines in sepsis: potent immunoregulators and potential therapeutic targets-an updated view. Mediators Inflamm. 2013;2013:165974. - PubMed
  40. Schroder K, Tschopp J. The inflammasomes. Cell. 2010;140:821–32. - PubMed
  41. Meziani F, Delabranche X, Asfar P, Toti F. Bench-to-bedside review: circulating microparticles-a new player in sepsis? Crit Care. 2010;14:236. - PubMed
  42. Martinod K, Wagner DD. Thrombosis: tangled up in NETs. Blood. 2014;123:2768–76. - PubMed
  43. 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. 2019;129:2163. - PubMed
  44. Huang W, Tang Y, Li L. HMGB1, a potent proinflammatory cytokine in sepsis. Cytokine. 2010;51:119–26. - PubMed
  45. Thomas JO, Stott K. H1 and HMGB1: modulators of chromatin structure. Biochem Soc Trans. 2012;40:341–6. - PubMed
  46. Stros M. HMGB proteins: interactions with DNA and chromatin. Biochim Biophys Acta. 2010;1799:101–13. - PubMed
  47. Teo SH, Grasser KD, Thomas JO. Differences in the DNA-binding properties of the HMG-box domains of HMG1 and the sex-determining factor SRY. Eur J Biochem. 1995;230:943–50. - PubMed
  48. Stros M. DNA bending by the chromosomal protein HMG1 and its high mobility group box domains. Effect of flanking sequences. J Biol Chem. 1998;273:10355–61. - PubMed
  49. Blair RH, Horn AE, Pazhani Y, Grado L, Goodrich JA, Kugel JF. The HMGB1 C-terminal tail regulates DNA bending. J Mol Biol. 2016;428:4060–72. - PubMed
  50. Scaffidi P, Misteli T, Bianchi ME. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature. 2002;418:191–5. - PubMed
  51. Ugrinova I, Pasheva E. HMGB1 protein: a therapeutic target inside and outside the cell. Adv Protein Chem Struct Biol. 2017;107:37–76. - PubMed
  52. Frank MG, Weber MD, Fonken LK, Hershman SA, Watkins LR, Maier SF. The redox state of the alarmin HMGB1 is a pivotal factor in neuroinflammatory and microglial priming: A role for the NLRP3 inflammasome. Brain Behav Immun. 2016;55:215–24. - PubMed
  53. 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;204:2913–23. - PubMed
  54. Venereau E, Casalgrandi M, Schiraldi M, Antoine DJ, Cattaneo A, De Marchis F, et al. Mutually exclusive redox forms of HMGB1 promote cell recruitment or proinflammatory cytokine release. J Exp Med. 2012;209:1519–28. - PubMed
  55. Yang H, Wang H, Ju Z, Ragab AA, Lundback P, Long W, et al. MD-2 is required for disulfide HMGB1-dependent TLR4 signaling. J Exp Med. 2015;212:5–14. - PubMed
  56. Kazama H, Ricci JE, Herndon JM, Hoppe G, Green DR, Ferguson TA. Induction of immunological tolerance by apoptotic cells requires caspase-dependent oxidation of high-mobility group box-1 protein. Immunity. 2008;29:21–32. - PubMed
  57. Gougeon ML, Bras M. Natural killer cells, dendritic cells, and the alarmin high-mobility group box 1 protein: a dangerous trio in HIV-1 infection? Curr Opin HIV AIDS. 2011;6:364–72. - PubMed
  58. Sha Y, Zmijewski J, Xu Z, Abraham E. HMGB1 develops enhanced proinflammatory activity by binding to cytokines. J Immunol. 2008;180:2531–7. - PubMed
  59. Urbonaviciute V, Furnrohr BG, Meister S, Munoz L, Heyder P, De Marchis F, et al. Induction of inflammatory and immune responses by HMGB1-nucleosome complexes: implications for the pathogenesis of SLE. J Exp Med. 2008;205:3007–18. - PubMed
  60. Zhao YF, Qiong Z, Zhang JF, Lou ZY, Zu HB, Wang ZG, et al. The synergy of aging and LPS exposure in a mouse model of Parkinson’s disease. Aging Dis. 2018;9:785–97. - PubMed
  61. Han R, Liu Z, Sun N, Liu S, Li L, Shen Y, et al. BDNF alleviates neuroinflammation in the hippocampus of type 1 diabetic mice via blocking the aberrant HMGB1/RAGE/NF-kappaB pathway. Aging Dis. 2019;10:611–25. - PubMed
  62. Li FJ, Zhang CL, Luo XJ, Peng J, Yang TL. Involvement of the MiR-181b-5p/HMGB1 pathway in Ang II-induced phenotypic transformation of smooth muscle cells in hypertension. Aging Dis. 2019;10:231–48. - PubMed
  63. Gardella S, Andrei C, Ferrera D, Lotti LV, Torrisi MR, Bianchi ME, et al. The nuclear protein HMGB1 is secreted by monocytes via a non-classical, vesicle-mediated secretory pathway. EMBO Rep. 2002;3:995–1001. - PubMed
  64. Schulman IG, Wang T, Wu M, Bowen J, Cook RG, Gorovsky MA, et al. Macronuclei and micronuclei in Tetrahymena thermophila contain high-mobility-group-like chromosomal proteins containing a highly conserved eleven-amino-acid putative DNA-binding sequence. Mol Cell Biol. 1991;11:166–74. - PubMed
  65. Zhang X, Wheeler D, Tang Y, Guo L, Shapiro RA, Ribar TJ, et al. Calcium/calmodulin-dependent protein kinase (CaMK) IV mediates nucleocytoplasmic shuttling and release of HMGB1 during lipopolysaccharide stimulation of macrophages. J Immunol. 2008;181:5015–23. - PubMed
  66. Bonaldi T, Talamo F, Scaffidi P, Ferrera D, Porto A, Bachi A, et al. Monocytic cells hyperacetylate chromatin protein HMGB1 to redirect it towards secretion. EMBO J. 2003;22:5551–60. - PubMed
  67. Deng M, Tang Y, Li W, Wang X, Zhang R, Zhang X, et al. The endotoxin delivery protein HMGB1 mediates caspase-11-dependent lethality in sepsis. Immunity. 2018;49:740–53 e7. - PubMed
  68. Lu B, Antoine DJ, Kwan K, Lundback P, Wahamaa H, Schierbeck H, et al. JAK/STAT1 signaling promotes HMGB1 hyperacetylation and nuclear translocation. Proc Natl Acad Sci USA. 2014;111:3068–73. - PubMed
  69. Rendon-Mitchell B, Ochani M, Li J, Han J, Wang H, Yang H, et al. IFN-gamma induces high mobility group box 1 protein release partly through a TNF-dependent mechanism. J Immunol. 2003;170:3890–7. - PubMed
  70. Dominguez Rubio AP, Correa F, Aisemberg J, Dorfman D, Bariani MV, Rosenstein RE, et al. Maternal administration of melatonin exerts short- and long-term neuroprotective effects on the offspring from lipopolysaccharide-treated mice. J Pineal Res. 2017;63:e12439. - PubMed
  71. Liu Z, Gan L, Xu Y, Luo D, Ren Q, Wu S, et al. Melatonin alleviates inflammasome-induced pyroptosis through inhibiting NF-kappaB/GSDMD signal in mice adipose tissue. J Pineal Res. 2017;63:e12414. - PubMed
  72. Rahim I, Djerdjouri B, Sayed RK, Fernandez-Ortiz M, Fernandez-Gil B, Hidalgo-Gutierrez A, et al. Melatonin administration to wild-type mice and nontreated NLRP3 mutant mice share similar inhibition of the inflammatory response during sepsis. J Pineal Res. 2017;63:e12410. - PubMed
  73. Magarinos AM, Schaafsma SM, Pfaff DW. Impacts of stress on reproductive and social behaviors. Front Neuroendocrinol. 2018;49:86–90. - PubMed
  74. Lee LC, Chen CM, Wang PR, Su MT, Lee-Chen GJ, Chang CY. Role of high mobility group box 1 (HMGB1) in SCA17 pathogenesis. PLoS One. 2014;9:e115809. - PubMed
  75. Hwang JS, Choi HS, Ham SA, Yoo T, Lee WJ, Paek KS, et al. Deacetylation-mediated interaction of SIRT1-HMGB1 improves survival in a mouse model of endotoxemia. Sci Rep. 2015;5:15971. - PubMed
  76. Stark K, Philippi V, Stockhausen S, Busse J, Antonelli A, Miller M, et al. Disulfide HMGB1 derived from platelets coordinates venous thrombosis in mice. Blood. 2016;128:2435–49. - PubMed
  77. Vogel S, Bodenstein R, Chen Q, Feil S, Feil R, Rheinlaender J, et al. Platelet-derived HMGB1 is a critical mediator of thrombosis. J Clin Invest. 2015;125:4638–54. - PubMed
  78. 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;38:40–8. - PubMed
  79. Tang Y, Zhao X, Antoine D, Xiao X, Wang H, Andersson U, et al. Regulation of posttranslational modifications of HMGB1 during immune responses. Antioxid Redox Signal. 2016;24:620–34. - PubMed
  80. Yang L, Xie M, Yang M, Yu Y, Zhu S, Hou W, et al. PKM2 regulates the Warburg effect and promotes HMGB1 release in sepsis. Nat Commun. 2014;5:4436. - PubMed
  81. Xie M, Yu Y, Kang R, Zhu S, Yang L, Zeng L, et al. PKM2-dependent glycolysis promotes NLRP3 and AIM2 inflammasome activation. Nat Commun. 2016;7:13280. - PubMed
  82. Kang R, Chen R, Zhang Q, Hou W, Wu S, Cao L, et al. HMGB1 in health and disease. Mol Asp Med. 2014;40:1–116. - PubMed
  83. Park A, Ra EA, Lee TA, Choi HJ, Lee E, Kang S, et al. HCMV-encoded US7 and US8 act as antagonists of innate immunity by distinctively targeting TLR-signaling pathways. Nat Commun. 2019;10:4670. - PubMed
  84. Volarevic V, Markovic BS, Jankovic MG, Djokovic B, Jovicic N, Harrell CR, et al. Galectin 3 protects from cisplatin-induced acute kidney injury by promoting TLR-2-dependent activation of IDO1/Kynurenine pathway in renal DCs. Theranostics. 2019;9:5976–6001. - PubMed
  85. Li T, Jiang S, Lu C, Yang W, Yang Z, Hu W, et al. Melatonin: another avenue for treating osteoporosis? J Pineal Res. 2019;66:e12548. - PubMed
  86. Chu KA, Wang SY, Yeh CC, Fu TW, Fu YY, Ko TL, et al. Reversal of bleomycin-induced rat pulmonary fibrosis by a xenograft of human umbilical mesenchymal stem cells from Wharton’s jelly. Theranostics. 2019;9:6646–64. - PubMed
  87. Yang H, Hreggvidsdottir HS, Palmblad K, Wang H, Ochani M, Li J, et al. A critical cysteine is required for HMGB1 binding to Toll-like receptor 4 and activation of macrophage cytokine release. Proc Natl Acad Sci USA. 2010;107:11942–7. - PubMed
  88. Park JS, Gamboni-Robertson F, He Q, Svetkauskaite D, Kim JY, Strassheim D, et al. High mobility group box 1 protein interacts with multiple Toll-like receptors. Am J Physiol Cell Physiol. 2006;290:C917–24. - PubMed
  89. Ramasamy R, Yan SF, Schmidt AM. Receptor for AGE (RAGE): signaling mechanisms in the pathogenesis of diabetes and its complications. Ann N Y Acad Sci. 2011;1243:88–102. - PubMed
  90. 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. 2019;130:1802. - PubMed
  91. Li T, Jiang S, Han M, Yang Z, Lv J, Deng C, et al. Exogenous melatonin as a treatment for secondary sleep disorders: a systematic review and meta-analysis. Front Neuroendocrinol. 2019;52:22–8. - PubMed
  92. Kokkola R, Andersson A, Mullins G, Ostberg T, Treutiger CJ, Arnold B, et al. RAGE is the major receptor for the proinflammatory activity of HMGB1 in rodent macrophages. Scand J Immunol. 2005;61:1–9. - PubMed
  93. Hori O, Brett J, Slattery T, Cao R, Zhang J, Chen JX, et al. The receptor for advanced glycation end products (RAGE) is a cellular binding site for amphoterin. Mediation of neurite outgrowth and co-expression of rage and amphoterin in the developing nervous system. J Biol Chem. 1995;270:25752–61. - PubMed
  94. LeBlanc PM, Doggett TA, Choi J, Hancock MA, Durocher Y, Frank F, et al. An immunogenic peptide in the A-box of HMGB1 protein reverses apoptosis-induced tolerance through RAGE receptor. J Biol Chem. 2014;289:7777–86. - PubMed
  95. Xu J, Jiang Y, Wang J, Shi X, Liu Q, Liu Z, et al. Macrophage endocytosis of high-mobility group box 1 triggers pyroptosis. Cell Death Differ. 2014;21:1229–39. - PubMed
  96. Yu H, Schwarzer K, Forster M, Kniemeyer O, Forsbach-Birk V, Straube E, et al. Role of high-mobility group box 1 protein and poly(ADP-ribose) polymerase 1 degradation in Chlamydia trachomatis-induced cytopathicity. Infect Immun. 2010;78:3288–97. - PubMed
  97. Park I, Kim M, Choe K, Song E, Seo H, Hwang Y, et al. Neutrophils disturb pulmonary microcirculation in sepsis-induced acute lung injury. Eur Respir J. 2019;53:1800786. - PubMed
  98. Neudecker V, Brodsky KS, Clambey ET, Schmidt EP, Packard TA, Davenport B, et al. Neutrophil transfer of miR-223 to lung epithelial cells dampens acute lung injury in mice. Sci Transl Med. 2017;9:eaah5360. - PubMed
  99. Zhang Y, Li X, Grailer JJ, Wang N, Wang M, Yao J, et al. Melatonin alleviates acute lung injury through inhibiting the NLRP3 inflammasome. J Pineal Res. 2016;60:405–14. - PubMed
  100. Brandenberger C, Kling KM, Vital M, Christian M. The role of pulmonary and systemic immunosenescence in acute lung injury. Aging Dis. 2018;9:553–65. - PubMed
  101. Sevransky JE, Martin GS, Shanholtz C, Mendez-Tellez PA, Pronovost P, Brower R, et al. Mortality in sepsis versus non-sepsis induced acute lung injury. Crit Care. 2009;13:R150. - PubMed
  102. Lan KC, Chao SC, Wu HY, Chiang CL, Wang CC, Liu SH, et al. Salidroside ameliorates sepsis-induced acute lung injury and mortality via downregulating NF-kappaB and HMGB1 pathways through the upregulation of SIRT1. Sci Rep. 2017;7:12026. - PubMed
  103. Wang Q, Wu X, Tong X, Zhang Z, Xu B, Zhou W. Xuebijing ameliorates sepsis-induced lung injury by downregulating HMGB1 and RAGE expressions in mice. Evid Based Complement Altern Med. 2015;2015:860259. - PubMed
  104. Wang SY, Li ZJ, Wang X, Li WF, Lin ZF. Effect of ulinastatin on HMGB1 expression in rats with acute lung injury induced by sepsis. Genet Mol Res. 2015;14:4344–53. - PubMed
  105. Li K, Yang J, Han X. Ketamine attenuates sepsis-induced acute lung injury via regulation of HMGB1-RAGE pathways. Int Immunopharmacol. 2016;34:114–28. - PubMed
  106. Zhou M, Fang H, Du M, Li C, Tang R, Liu H, et al. The modulation of regulatory T cells via HMGB1/PTEN/beta-catenin axis in LPS induced acute lung injury. Front Immunol. 2019;10:1612. - PubMed
  107. 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;9:37–45. - PubMed
  108. Xu W, Lu Y, Yao J, Li Z, Chen Z, Wang G, et al. Novel role of resveratrol: suppression of high-mobility group protein box 1 nucleocytoplasmic translocation by the upregulation of sirtuin 1 in sepsis-induced liver injury. Shock. 2014;42:440–7. - PubMed
  109. Zheng S, Pan Y, Wang C, Liu Y, Shi M, Ding G. HMGB1 turns renal tubular epithelial cells into inflammatory promoters by interacting with TLR4 during sepsis. J Interferon Cytokine Res. 2016;36:9–19. - PubMed
  110. Yang X, Cheng X, Tang Y, Qiu X, Wang Z, Fu G, et al. The role of type 1 interferons in Gram-negative bacteria-induced coagulation. Blood. 2020;135:1087–100. - PubMed
  111. Zhao F, Fang Y, Deng S, Li X, Zhou Y, Gong Y, et al. Glycyrrhizin protects rats from sepsis by blocking HMGB1 signaling. Biomed Res Int. 2017;2017:9719647. - PubMed
  112. Gil M, Kim YK, Hong SB, Lee KJ. Naringin decreases TNF-alpha and HMGB1 release from LPS-stimulated macrophages and improves survival in a CLP-induced sepsis mice. PLoS One. 2016;11:e0164186. - PubMed
  113. van Zoelen MA, Laterre PF, van Veen SQ, van Till JW, Wittebole X, Bresser P, et al. Systemic and local high mobility group box 1 concentrations during severe infection. Crit Care Med. 2007;35:2799–804. - PubMed
  114. Sunden-Cullberg J, Norrby-Teglund A, Rouhiainen A, Rauvala H, Herman G, Tracey KJ, et al. Persistent elevation of high mobility group box-1 protein (HMGB1) in patients with severe sepsis and septic shock. Crit Care Med. 2005;33:564–73. - PubMed
  115. Karlsson S, Pettila V, Tenhunen J, Laru-Sompa R, Hynninen M, Ruokonen E. HMGB1 as a predictor of organ dysfunction and outcome in patients with severe sepsis. Intensive Care Med. 2008;34:1046–53. - PubMed
  116. Fu GX, Chen AF, Zhong Y, Zhao J, Gu YJ. Decreased serum level of HMGB1 and MyD88 during human aging progress in healthy individuals. Aging Clin Exp Res. 2016;28:175–80. - PubMed
  117. Hatada T, Wada H, Nobori T, Okabayashi K, Maruyama K, Abe Y, et al. Plasma concentrations and importance of High Mobility Group Box protein in the prognosis of organ failure in patients with disseminated intravascular coagulation. Thromb Haemost. 2005;94:975–9. - PubMed
  118. Bianchi ME, Crippa MP, Manfredi AA, Mezzapelle R, Rovere Querini P, Venereau E. High-mobility group box 1 protein orchestrates responses to tissue damage via inflammation, innate and adaptive immunity, and tissue repair. Immunol Rev. 2017;280:74–82. - PubMed
  119. Schierbeck H, Pullerits R, Pruunsild C, Fischer M, Holzinger D, Laestadius A, et al. HMGB1 levels are increased in patients with juvenile idiopathic arthritis, correlate with early onset of disease, and are independent of disease duration. J Rheumatol. 2013;40:1604–13. - PubMed
  120. Horiuchi T, Sakata N, Narumi Y, Kimura T, Hayashi T, Nagano K, et al. Metformin directly binds the alarmin HMGB1 and inhibits its proinflammatory activity. J Biol Chem. 2017;292:8436–46. - PubMed

Publication Types