Damage‐associated molecular patterns: Their impact on the liver and beyond during acetaminophen overdose Damage-Associated Molecular Patterns: Their Impact on the Liver and Beyond During Acetaminophen Overdose See Article on Page 1971 A cetaminophen (APAP) is one of the most commonly used drugs for treating pain and fever. Although it is safe at therapeutic dosage levels, overdose of APAP causes severe liver injury (APAP- induced liver injury; AILI), with the potential to progress to liver failure. Up to 40% of patients who suffer from liver failure will die (or undergo liver transplant).1 Data combined from 22 specialty medical centers in the United States revealed that AILI accounts for approxi- mately half of acute liver failure cases and results in more than 56,000 emergency room visits, 2,600 hospitaliza- tions, and an estimated 458 deaths each year.1 APAP hepatotoxicity is initiated by the generation of a chemically reactive metabolite (N-acetyl-p-benzo- quinone imine; NAPQI). NAPQI depletes liver gluta- thione and covalently binds to cellular proteins, thereby causing mitochondrial dysfunction.2-6 Studies using rodent models in the last four decades have demonstrated that mitochondrial disruption is the key underlying mechanism of AILI (Fig. 1FIG1)). Cova- lent binding to mitrochondrial proteins by NAPQI causes oxidative stress and mitochondrial membrane permeability transition pore opening, which triggers the collapse of membrane potential, cessation of aden- osine triphosphate (ATP) production, and the release of apoptosis-inducing factor and endonuclease G.7-11 Together, the mitochondrial dysfunction, energy crisis, and nuclear DNA damage result in hepatocyte necrosis. In recent years, there has been a growing interest to investigate whether downstream events of early hepato- cyte necrosis contribute to the aggravation and pro- gression of AILI. Necrotic cells release a number of damage-associated molecular pattern (DAMP) mole- cules, such as high-mobility group box-1, heat-shock proteins, hyaluronan, fibronectin, cardiolipin, and DNA fragments. Upon activation by DAMP mole- cules, innate immune cells infiltrate the damaged area and release cytokines and cheomokines, thereby caus- ing tissue sterile inflammation.12-22 The soluble prod- ucts of innate immune cells can exacerbate tissue dam- age, as well as promote wound healing (Fig. 1). Perhaps as a result of this dichotomy, the overall con- tribution of innate immune cells, such as neutrophils, to AILI remains unclear. In this issue of HEPATOLOGY, Marques et al.23 report on a study demonstrating that DAMP molecules released from necrotic hepatocytes recruit and activate neutrophils in the liver, which, in turn, amplify AILI. Three experimental approaches were employed to elu- cidate the pathological role of neutrophils in AILI. Consistent with published reports,24,25 the present study shows that neutrophil depletion by an anti-Gr-1 antibody significantly attenuates AILI. Furthermore, the combined use of a CXC chemokine receptor 2 an- tagonist and a formyl peptide receptor 1 (FPR1) an- tagonist also blocks hepatic recruitment of neutrophils and mitigates AILI. This approach is based on the investigators’ previous finding that an intravascular gra- dient of chemokines and mitochondria-derived formyl peptides collaboratively guide neutrophils to sites of liver necrosis.26 These two separate in vivo studies demonstrate that liver injury initiated by APAP chal- lenge is amplified by infiltrating neutrophils. The investigators further examined the cytotoxic potential of neutrophils against hepatocytes. Neutrophils isolated from healthy individuals were cocultured with APAP- treated HepG2 cells. Data show that the cytotoxicity of HepG2 cells is enhanced by neutrophils in a cell- contact–dependent manner, and that necrotic HepG2 cells significantly increase reactive oxygen species pro- duction by netrophils. Collectively, these findings pro- vide evidence to support a pathological role of neutro- phils during AILI. Traumatic injury is known to cause ‘‘septic-like’’ sys- temic inflammatory response in the absence of infec- tion.27 The underlying mechanism is recently Abbreviations: AILI, APAP-induced liver injury; APAP, acetaminophen; ATP, adenosine triphosphate; DAMP, damage-associated molecular pattern; FPR1, formyl peptide receptor 1; IL, interleukin; mtDNA, mitrochondrial DNA; NAPQI, N-acetyl-p-benzoquinone imine; TLR-9, Toll-like receptor 9. Address reprint requests to: Cynthia Ju, Ph.D., School of Pharmacy, University of Colorado Health Sciences Center, 4200 East 9th Avenue, C238, Denver, CO 80246. E-mail: Cynthia.ju@uchsc.edu; fax: 303-724-7266. Copyright VC 2012 by the American Association for the Study of Liver Diseases. View this article online at wileyonlinelibrary.com. DOI 10.1002/hep.25920 Potential conflict of interest: Nothing to report. 1599 elucidated by the detection of mitrochondrial DNA (mtDNA) and formyl peptides released in the serum of trauma patients.28 Circulating mitochondrial DAMPs activate neutrophils through Toll-like receptor 9 (TLR-9) and FPR1, respectively, thereby eliciting neutrophil-mediated organ injury.28 Because APAP causes mitochrondria damage, it is likely that mito- chondrial contents are released into the circulation. Evidence supporting this hypothesis is provided by a recent study detecting circulating mitochondrial bio- markers, including mtDNA and glutamate dehydro- genase, in the serum of patients with AILI.29 Similarly, the present study shows a significant increase in serum mtDNA levels in acute liver failure patients, compared to healthy volunteers. The elevation of circulating mtDNA is also observed in APAP-treated mice. The effect of mtDNA release on AILI is revealed by a sig- nificant decrease of liver injury in TLR-9�/� mice, compared to wild-type mice. This finding is consistent with a published report of protection against AILI by TLR-9 antagonists and in TLR-9�/� mice.30 APAP-induced liver failure is accompanied by other tissue complications, such as encephalopathy, coagul- opathy, renal failure, metabolic derangements, cardio- vascular compromises, and severe lung injury.31,32 Toxic effects on remote tissues may be the result of the original insult of APAP, but, in many cases, are conse- quences of severe liver injury. A novel finding of the present study is the observation of marked lung injury in mice treated with APAP. Data suggest that lung injury is the result of the systemic release of mitochon- drial DAMPs, because blockade of FPR1 or deletion of TLR-9 significantly reduced lung injury. From their results, the investigators summarize a mechanistic model for APAP-induced liver damage and lung inflammation. APAP-initiated hepatocyte necrosis causes the release of mitochondrial DAMPs and che- mokines, which lead to hepatic recruitment of neutro- phils that amplify liver damage. The release of mito- chondrial DAMPs also triggers systemic inflammation and causes organ injury at remote sites. Many studies clearly support that neutrophils are recruited into the liver after cellular damage initiated by APAP challenge. However, the key unresolved ques- tion is whether or not the infiltrated neutrophils are activated and aggravate AILI. Data from some studies, including the present one, provide evidence for a path- ological role of neutrophils in AILI. However, other studies have demonstrated that (1) neutrophils recruited into the liver are not activated,33 (2) blocking neutro- phil recruitment does not affect AILI,33,34 and (3) even activating neutrophils by endotoxin or interleukin (IL)- 1b does not worsen AILI.33,35 Aside from the dichot- omy of tissue-damaging and -repair functions of neu- trophils, these discrepancies can be, at least in part, explained by different experimental protocols employed by various research groups. For example, two critical experimental conditions that can significantly affect the severity and kinetics of AILI include the mouse strains, as well as the dose and route of administration of APAP. Therefore, direct comparisons can only be made when the experimental approaches are unified. CYNTHIA JU, PH.D. School of Pharmacy University of Colorado Health Sciences Center Denver, CO References 1. Larson AM, Polson J, Fontana RJ, Davern TJ, Lalani E, Hynan LS, et al. Acetaminophen-induced acute liver failure: results of a United States mul- ticenter, prospective study. HEPATOLOGY 2005;42:1364-1372. 2. Jollow DJ, Mitchell JR, Potter WZ, Davis DC, Gillette JR, Brodie BB. Acetaminophen-induced hepatic necrosis. II. Role of covalent binding in vivo. J Pharmacol Exp Ther 1973;187:195-202. Fig. 1. Mechanism of AILI. Metabolic activation of APAP generates NAPQI. Glutathione depletion and protein binding by NAPQI cause mi- tochondrial oxidative stress, mitochondrial permeability transition (MPT), ATP depletion, and release of apoptosis-inducing factor (AIF) and endonuclease G (ENDOG), all of which culminate in hepatocyte necrosis. DAMPs are released and trigger the activation of innate immune cells, such as neutrophils (PMN), Kupffer cells (KC), and nat- ural killer (NK) and NK T cells. Some soluble factors produced by acti- vated innate immune cells can cause further tissue damage, such as tumor necrosis factor (TNF)-a, IL-1b, interferon (IFN)-c, and macro- phage migration inhibitory factor (MIF). However, the same factors (TNF-a and IL-1b) and other mediators, including IL-6, IL-4, IL-13, IL- 10, and vascular endothelial growth factor (VEGF), are important in promoting liver repair. 1600 JU HEPATOLOGY, November 2012 3. Mitchell JR, Jollow DJ, Potter WZ, Gillette JR, Brodie BB. Acetamino- phen-induced hepatic necrosis. IV. Protective role of glutathione. J Pharmacol Exp Ther 1973;187:211-217. 4. Qiu Y, Benet LZ, Burlingame AL. Identification of the hepatic protein targets of reactive metabolites of acetaminophen in vivo in mice using two-dimensional gel electrophoresis and mass spectrometry. J Biol Chem 1998;273:17940-17953. 5. Qiu Y, Benet LZ, Burlingame AL. Identification of hepatic protein tar- gets of the reactive metabolites of the non-hepatotoxic regioisomer of acetaminophen, 30-hydroxyacetanilide, in the mouse in vivo using two- dimensional gel electrophoresis and mass spectrometry. Adv Exp Med Biol 2001;500:663-673. 6. Tirmenstein MA, Nelson SD. Subcellular binding and effects on cal- cium homeostasis produced by acetaminophen and a nonhepatotoxic regioisomer, 30-hydroxyacetanilide, in mouse liver. J Biol Chem 1989; 264:9814-9819. 7. Jaeschke H. Glutathione disulfide formation and oxidant stress during acetaminophen-induced hepatotoxicity in mice in vivo: the protective effect of allopurinol. J Pharmacol Exp Ther 1990;255:935-941. 8. Jaeschke H, Knight TR, Bajt ML. The role of oxidant stress and reac- tive nitrogen species in acetaminophen hepatotoxicity. Toxicol Lett 2003;144:279-288. 9. Jaeschke H, Bajt ML. Intracellular signaling mechanisms of acetamino- phen-induced liver cell death. Toxicol Sci 2006;89:31-41. 10. Meyers LL, Beierschmitt WP, Khairallah EA, Cohen SD. Acetamino- phen-induced inhibition of hepatic mitochondrial respiration in mice. Toxicol Appl Pharmacol 1988;93:378-387. 11. Ramsay RR, Rashed MS, Nelson SD. In vitro effects of acetaminophen metabolites and analogs on the respiration of mouse liver mitochon- dria. Arch Biochem Biophys 1989;273:449-457. 12. Asea A, Rehli M, Kabingu E, Boch JA, Bare O, Auron PE, et al. Novel signal transduction pathway utilized by extracellular HSP70: role of toll-like receptor (TLR) 2 and TLR4. J Biol Chem 2002;277: 15028-15034. 13. Okamura Y, Watari M, Jerud ES, Young DW, Ishizaka ST, Rose J, et al. The extra domain A of fibronectin activates Toll-like receptor 4. J Biol Chem 2001;276:10229-10233. 14. Peitsch MC, Tschopp J, Kress A, Isliker H. Antibody-independent acti- vation of the complement system by mitochondria is mediated by car- diolipin. Biochem J 1988;249:495-500. 15. Shi Y, Evans JE, Rock KL. Molecular identification of a danger signal that alerts the immune system to dying cells. Nature 2003;425: 516-521. 16. Wallin RP, Lundqvist A, Moré SH, von Bonin A, Kiessling R, Ljungg- ren HG. Heat-shock proteins as activators of the innate immune sys- tem. Trends Immunol 2002;23:130-135. 17. 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. Sci- ence 1999;285:248-251. 18. Martin-Murphy BV, Holt MP, Ju C. The role of damage associated molecular pattern molecules in acetaminophen-induced liver injury in mice. Toxicol Lett 2010;192:387-394. 19. Scaffidi P, Misteli T, Bianchi ME. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature 2002;418: 191-195. 20. Termeer C, Benedix F, Sleeman J, Fieber C, Voith U, Ahrens T, et al. Oligosaccharides of Hyaluronan activate dendritic cells via toll-like re- ceptor 4. J Exp Med 2002;195:99-111. 21. Lamphier MS, Sirois CM, Verma A, Golenbock DT, Latz E. TLR9 and the recognition of self- and non-self nucleic acids. Ann N Y Acad Sci 2006;1082:31-43. 22. Jahr S, Hentze H, Englisch S, Hardt D, Fackelmayer FO, Hesch RD, Knippers R. DNA fragments in the blood plasma of cancer patients: quantitations and evidence for their origin from apoptotic and necrotic cells. Cancer Res 2001;61:1659-1665. 23. Marques PE, Amaral SS, Pires DA, Noqueira LL, Soriani A, Freire Lima BH, et al. Chemokines and mitochondrial products activate neu- trophils to amplify organ injury during mouse acute liver failure. HEPA- TOLOGY 2012;56:1971-1982. 24. Ishida Y, Kondo T, Kimura A, Tsuneyama K, Takayasu T, Mukaida N. Opposite roles of neutrophils and macrophages in the pathogenesis of acetaminophen-induced acute liver injury. Eur J Immunol 2006;36: 1028-1038. 25. Liu ZX, Han D, Gunawan B, Kaplowitz N. Neutrophil depletion pro- tects against murine acetaminophen hepatotoxicity. HEPATOLOGY 2006; 43:1220-1230. 26. McDonald B, Pittman K, Menezes GB, Hirota SA, Slaba I, Waterhouse CC, et al. Intravascular danger signals guide neutrophils to sites of ster- ile inflammation. Science 2010;330:362-366. 27. Bone RC. Toward an epidemiology and natural history of SIRS (sys- temic inflammatory response syndrome). JAMA 1992;268:3452-3455. 28. Zhang Q, Raoof M, Chen Y, Sumi Y, Sursal T, Junger W, et al. Circu- lating mitochondrial DAMPs cause inflammatory responses to injury. Nature 2010;464:104-107. 29. McGill MR, Sharpe MR, Williams CD, Taha M, Curry SC, Jaeschke H. The mechanism underlying acetaminophen-induced hepatotoxicity in humans and mice involves mitochondrial damage and nuclear DNA fragmentation. J Clin Invest 2012;122:1574-1583. 30. Imaeda AB, Watanabe A, Sohail MA, Mahmood S, Mohamadnejad M, Sutterwala FS, et al. Acetaminophen-induced hepatotoxicity in mice is dependent on Tlr9 and the Nalp3 inflammasome. J Clin Invest 2009; 119:305-314. 31. Craig DG, Bates CM, Davidson JS, Martin KG, Hayes PC, Simpson KJ. Staggered overdose pattern and delay to hospital presentation are associated with adverse outcomes following paracetamol-induced hepa- totoxicity. Br J Clin Pharmacol 2012;73:285-294. 32. Baudouin SV, Howdle P, O’Grady JG, Webster NR. Acute lung injury in fulminant hepatic failure following paracetamol poisoning. Thorax 1995;50:399-402. 33. Williams CD, Bajt ML, Farhood A, Jaeschke H. Acetaminophen- induced hepatic neutrophil accumulation and inflammatory liver injury in CD18-deficient mice. Liver Int 2010;30:1280-1292. 34. Cover C, Liu J, Farhood A, Malle E, Waalkes MP, Bajt ML, Jaeschke H. Pathophysiological role of the acute inflammatory response during acetaminophen hepatotoxicity. Toxicol Appl Pharmacol 2006;216: 98-107. 35. Williams CD, Farhood A, Jaeschke H. Role of caspase-1 and interleu- kin-1beta in acetaminophen-induced hepatic inflammation and liver injury. Toxicol Appl Pharmacol 2010;247:169-178. HEPATOLOGY, Vol. 56, No. 5, 2012 JU 1601