Skip to main content

Heme oxygenase-1 induction in hepatocytes and non-parenchymal cells protects against liver injury during endotoxemia

Abstract

Introduction

Heme oxygenase-1 (HO-1) is a stress response enzyme, which catalyses the breakdown of heme into biliverdin-IX alpha, carbon monoxide and ferrous iron. Under situations of oxidative stress, heat stress, ischemia/reperfusion injury or endotoxemia, HO-1 has been shown to be induced and to elicit a protective effect. The mechanism of how this protective effect is executed is unknown.

Results

HO-1 induction with cobalt protoporphorin (Co-PP) dose-dependently protected against apoptotic cell death as well as neutrophil-mediated oncosis in the galactosamine/endotoxin (Gal/ET) shock model. Induction of HO-1 with Co-PP dose-dependently protected against neutrophil-mediated oncosis as indicated by attenuated ALT release and TNF-mediated apoptotic cell death as indicated by reduced caspase-3 activation. HO-1 induction did not attenuate Gal/ET-induced TNF-alpha formation. Furthermore, a similar protective effect with the high dose of Co-PP was observed when animals were treated with Gal/TNF-alpha.

Conclusions

HO-1 induction attenuates apoptosis and neutrophil-mediated oncosis in the Gal/ET shock model. However, the protective effect is not due to the reduction of TNF-alpha release or the attenuation of neutrophil accumulation in the liver sinusoids.

Introduction

Heme oxygenase (HO) catalyzes the oxidative cleavage of Fe-protoporphyrin-IX yielding equimolar amounts of biliverdin-IX alpha, free divalent iron, and carbon monoxide (CO) [1]. Among the three isoenzymes cloned to date, only heme oxygenase-1 (HO-1) can be induced by a variety of disparate stimuli, most of which are linked by their ability to provoke oxidative stress [1]. Induction of HO-1 may protect the cell against oxidative injury by a) controlling intracellular levels of "free" heme (a prooxidant), b) producing biliverdin (an antioxidant), c) improving nutritive perfusion via CO release, and d) fostering the synthesis of the Fe-binding protein ferritin [1]. In the liver, HO-1 induction protected against ischemia/reperfusion injury [2, 3] and endotoxemia [4]. However, the mechanism of protection is unclear. In particular, it is controversial whether HO-1 induction in the liver protects against apoptotic and/or oncotic cell death. To address this question, we investigated the beneficial effect of HO-1 induction in the galactosamine/endotoxin (Gal/ET) shock model. Cell injury in this model involves TNF-induced apoptosis [5] as well as a neutrophil-mediated oncosis, which is caused mainly by reactive oxygen species [6, 7].

Methods

Male C3Heb/FeJ mice (Jackson Laboratories, Bar Harbor, ME) were treated i.p. with 700 mg/kg D-galactosamine (Sigma Chemical Co., St. Louis, MO) in combination with 100 micrograms/kg Salmonella abortus equi endotoxin (Gal/ET) or 20 micrograms/kg murine TNF-alpha (Gal/TNF). Some animals were pretreated with 5 or 15 mg/kg of cobalt-protoporphyrin (Alexis, San Diego, CA) 18 h before Gal/ET. The following parameters were measured as previously described: caspase-3 activity [5], caspase-3 processing and HO-1 expression [8], plasma ALT activities [5], plasma TNF-alpha [7], imunohistochemistry [9], neutrophil staining and histological assessment of necrosis [10].

Results and Discussion

Western blot and immunohistochemical analysis indicated that cobalt protoporphyrin (Co-PP) dose-dependently induced HO-1 expression in hepatocytes and nonparenchymal cells, especially Kupffer cells. To investigate if increased HO-1 expression affected apoptotic or oncotic cell death, animals were treated with galactosamine/endotoxin (Gal/ET). It was previously shown that Gal/ET treatment induces TNF-alpha formation, which activates and recruits neutrophils into the liver vasculature [6] and causes a caspase-dependent parenchymal cell apoptosis [5]. In addition, the apoptotic cell death triggers neutrophil extravasation with massive aggravation of the apoptotic cell injury [5]. Gal/ET treatment caused a 10-fold increase of caspase-3 activity (indicator of apoptosis) together with substantial ALT release into the plasma (indicator of oncosis) (Table 1). Overall cell death (necrosis) was estimated to be 42 – 4%. Caspase-3 activity was reduced by 40% (5 mg/kg CoPP) or 90% (15 mg/kg Co-PP). On the other hand, ALT activities were reduced to values not different from baseline (Table 1). Neutrophil accumulation in liver sinusoids and parenchyma (174 – 10 neutrophils/50 high power fields) and adherence in venules (30 – 3 neutrophils/10 venules) were not affected by Co-PP. Furthermore, plasma TNF-alpha levels (11.6 – 1.2 ng/ml) were not reduced by Co-PP administration. These data suggest that HO-1 induction reduced both apoptotic and oncotic cell death but did not prevent production of key inflammatory mediators or neutrophil recruitment. At the low dose of Co-PP, the inhibition of neutrophil-mediated oncosis was more effective compared to attenuation of apoptosis.

Table 1 Hepatoprotection by Heme Oxygenase-1 Induction

To investigate if the beneficial effect was due to HO-1 induction, animals were additionally treated with the HO-1 inhibitor tin-protoporphyrin (Sn-PP). Treatment with Sn-PP alone did not affect liver injury after Gal/ET (Table 1). However, the beneficial effect of Co-PP treatment on Gal/ET-mediated apoptosis as well as neutrophil-induced oncosis was completely reversed by Sn-PP (Table 1). These data suggest that the hepatoprotection observed with Co-PP administration was mainly due to HO-1 induction.

Although the TNF response after Gal/ET could not explain the protective effect of Co-PP, the potential protection of HO-1 induction against Gal/TNF-induced liver injury was investigated. Gal/TNF caused massive caspase-3 activation (Table 1), which was confirmed by evaluation of procaspase-3 processing (data not shown). In addition, the increase in plasma ALT activities indicates oncotic cell injury. Overall, 40 – 3% of all hepatocytes were necrotic at 7 h after Gal/TNF administration. The low dose of Co-PP had no effect on any of the parameters measured. However, the high dose of Co-PP attenuated caspase-3 activities by 45% and plasma ALT activities by >90% (Table 1). The total number of necrotic cells, which were mainly from apoptotic cells, was reduced to 10 – 2%. These data suggest that the high dose of Co-PP partially reduced apoptosis but completely eliminated oncotic necrosis. In this model, hepatocellular apoptosis triggers neutrophil extravasation and cytotoxicity [5]. Elimination of apoptotic cell injury with a caspase inhibitor completely prevented neutrophil-induced liver injury [5]. In two different experiments, apoptosis was only partially prevented but oncotic cell death was almost completely eliminated by Co-PP treatment. This indicates that HO-1 induction affected both forms of cell death.

Unexpectedly, the massive HO-1 induction with increased formation of antioxidants in Kupffer cells did not attenuate TNF formation. In contrast, high doses of the antioxidant dimethyl sulfoxide eliminated TNF formation [11] and glutathione peroxidase deficiency enhanced TNF generation after Gal/ET [7]. In addition, HO-1 induction was also effective after Gal/TNF administration. Together, these data suggest that HO-1 induction in hepatocytes was more critical for the protective effect than the quantitatively higher increase of HO-1 levels in non-parenchymal cells. Moreover, an effect on hepatocellular antioxidant levels is unlikely the mechanism of protection. Recent data indicate that CO may be the more relevant mediator of the anti-apoptotic and cytoprotective effects of HO-1 induction [12, 13].

Conclusions

Induction of HO-1 in non-parenchymal cells and in hepatocytes with Co-PP dose-dependently protected against TNF-mediated apoptotic cell death and subsequent neutrophil-induced injury to hepatocytes. The protection is not due to inhibition of TNF-alpha formation or attenuation of neutrophil activation. HO-1 induction increases the resistance to both forms of cell death in hepatocytes.

References

  1. Bauer M, Bauer I: Hemeoxygenase-1: redoxregulation and the role in the hepatic response to oxidative stress. Antioxid Redox Signal. 2002, 4: 749-758. 10.1089/152308602760598891.

    Article  CAS  PubMed  Google Scholar 

  2. Rensing H, Bauer I, Datene V, Patau C, Pannen BH, Bauer M: Differential expression pattern of heme oxygenase-1/heat shock protein 32 and nitric oxide synthase-II and their impact on liver injury in a rat model of hemorrhage and resuscitation. Crit Care Med. 1999, 27: 2766-2775. 10.1097/00003246-199912000-00027.

    Article  CAS  PubMed  Google Scholar 

  3. Amersi F, Buelow R, Kato H, Ke B, Coito AJ, Shen XD, Zhao D, Zaky J, Melinek J, Lassman CR, et al: Upregulation of heme oxygenase-1 protects genetically fat Zucker rat livers from ischemia/reperfusion injury. J Clin Invest. 1999, 104: 1631-1639. 10.1172/JCI7903.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  4. Kyokane T, Norimizu S, Taniai H, Yamaguchi T, Takeoka S, Tsuchida E, Naito M, Nimura Y, Ishimura Y, Suematsu M: Carbon monoxide from heme catabolism protects against hepatobiliary dysfunction in endotoxin-treated rat liver. Gastroenterology. 2001, 120: 1227-1240. 10.1053/gast.2001.23249.

    Article  CAS  PubMed  Google Scholar 

  5. Jaeschke H, Fisher MA, Lawson JA, Simmons CA, Farhood A, Jones DA: Activation of caspase-3 (CPP32)-like proteases is essential for TNF-alpha-induced hepatic parenchymal cell apoptosis and neutrophil-mediated necrosis in a murine endotoxin shock model. J Immunol. 1998, 160: 3480-3486.

    CAS  PubMed  Google Scholar 

  6. Jaeschke H, Farhood A, Smith CW: Neutrophil-induced liver cell injury in endotoxin shock is a CD11b/CD18-dependent mechanism. Am J Physiol. 1991, 261: G1051-G1056.

    CAS  PubMed  Google Scholar 

  7. Jaeschke H, Ho Y-S, Fisher MA, Lawson JA, Farhood A: Glutathione peroxidase deficient mice are more susceptible to neutrophil-mediated hepatic parenchymal cell injury during endotoxemia: importance of an intracellular oxidant stress. Hepatology. 1999, 29: 443-450. 10.1002/hep.510290222.

    Article  CAS  PubMed  Google Scholar 

  8. Bajt ML, Lawson JA, Vonderfecht SL, Gujral JS, Jaeschke H: Protection against Fas receptor-mediated apoptosis in hepatocytes and nonparenchymal cells by a caspase-8 inhibitor in vivo: evidence for postmitochondrial processing of caspase-8. Toxicol Sci. 2000, 58: 109-117. 10.1093/toxsci/58.1.109.

    Article  CAS  PubMed  Google Scholar 

  9. Knight TR, Kurtz A, Bajt ML, Hinson JA, Jaeschke H: Vascular and hepatocellular peroxynitrite formation during acetaminophen-induced liver injury: role of mitochondrial oxidant stress. Toxicol Sci. 2001, 62: 212-220. 10.1093/toxsci/62.2.212.

    Article  CAS  PubMed  Google Scholar 

  10. Jaeschke H, Farhood A, Fisher MA, Smith CW: Sequestration of neutrophils in the hepatic vasculature during endotoxemia is independent of beta2 integrins and intercellular adhesion molecule-1. Shock. 1996, 6: 351-356.

    Article  CAS  PubMed  Google Scholar 

  11. Essani NA, Fisher MA, Jaeschke H: Inhibition of NF-kappa B activation by dimethyl sulfoxide correlates with suppression of TNF-alpha formation, reduced ICAM-1 gene transcription and protection against endotoxin-induced liver injury. Shock. 1997, 7: 90-96. 10.1097/00024382-199702000-00003.

    Article  CAS  PubMed  Google Scholar 

  12. Otterbein LE, Mantell LL, Choi AM: Carbon monoxide provides protection against hyperoxic lung injury. Am J Physiol. 1999, 276: L688-L694.

    CAS  PubMed  Google Scholar 

  13. Brouard S, Otterbein LE, Anrather J, Tobiasch E, Bach FH, Choi AM, Soares MP: Carbon monoxide generated by hemoxygenase-1 suppresses endothelial cell apoptosis. J Exp Med. 2000, 192: 1015-1026. 10.1084/jem.192.7.1015.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Hartmut Jaeschke.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Dorman, R.B., Bajt, M.L., Farhood, A. et al. Heme oxygenase-1 induction in hepatocytes and non-parenchymal cells protects against liver injury during endotoxemia. Comp Hepatol 3 (Suppl 1), S42 (2004). https://doi.org/10.1186/1476-5926-2-S1-S42

Download citation

  • Published:

  • DOI: https://doi.org/10.1186/1476-5926-2-S1-S42

Keywords