The effects of early and late administration of inhibitors of inducible nitric oxide synthase in a thioacetamide-induced model of acute hepatic failure in the rat

https://doi.org/10.1016/S0168-8278(03)00050-3Get rights and content

Abstract

Background/Aims: Nitric oxide (NO) is a pivotal mediator of inflammation. Its role in acute hepatic failure (AHF) is controversial. We investigated the role of NO, and the hypothesis that inhibition of inducible NO synthase (iNOS) activity would improve outcome in liver failure in rats, using the iNOS inhibitors l-NAME and aminoguanidine (AMG).

Methods: AHF was induced by two intraperitoneal injections of thioacetamide (TAA). Seven groups (n=10) were studied. Group I: TAA alone. Groups II, III and IV were additionally pre-treated with the NO precursor l-arginine (300 mg/kg i.p.), or iNOS inhibitors AMG (100 mg/kg s.c.), or NG-nitro-l-arginine methyl ester (l-NAME) (100 mg/kg s.c.) for 5 days, respectively. Groups V, VI and VII received l-arginine, AMG or l-NAME commencing immediately after TAA administration. Clinical and biochemical parameters were assessed serially, and mortality investigated in further similar cohorts for each regime.

Results: AMG, pre-treatment but not post-treatment, significantly improved outcome including mortality (10 vs. 70%, P<0.005). The less selective iNOS inhibitor l-NAME was not beneficial. Arginine pre-and post-treatment, and iNOS inhibition post-treatment, worsened clinical parameters of TAA-induced liver failure.

Conclusions: Administration of the iNOS inhibitor AMG prior to insult reduces the severity of damage and improves mortality.

Introduction

Nitric oxide (NO) is a highly reactive oxidant. In the liver it is produced both by parenchymal and non-parenchymal cells. Over-production of NO in the liver from l-arginine via inducible nitric oxide synthase (iNOS) [1], [2], [3] has been implicated as an important part of the cascade of events that takes place in the pathogenesis of septic shock and in various forms of hepatic injury, inflammation and acute hepatic failure (AHF) [4], [5]. Supplementation of the NO precursor l-arginine has been shown to exacerbate damage in models of inflammation and injury [6].

The precise mechanisms underlying the actions of NO are controversial and as yet are unknown. NO has been implicated in the haemodynamic changes in the systemic circulation of patients with both AHF and chronic liver disease [7]. It is as an important contributor to haemodynamic changes in the brain and in the pathogenesis of cerebral oedema associated with AHF [8], [9]. In vitro and in vivo studies have also shown NO to down-regulate cytochrome P450 and suppress liver protein and DNA synthesis, and to induce apoptosis and necrosis, all of which may contribute to liver failure [10], [11]. NO also inhibits catalase activity, suggesting that it may alter the detoxification of cytotoxic free radicals, and reacts with superoxide anions to form peroxynitrite, which can react with sulphydryl residues in cell membranes leading to lipid peroxidation, and with DNA leading to cytotoxicity [2].

As modulators of NO production have become available, it is important to assess their potential utility in AHF. We utilized the thioacetamide (TAA) model of AHF in the rat. Thioacetamide is metabolized to toxic metabolites thioacetamide sulfine (sulphoxide) and sulfene (sulphone) [12], [13]. Damage involves generation of free radicals and superoxide anions causing lipid peroxidation [14]. Cellular consequences include interruption of cellular calcium homeostasis, disruption of membranes of the endoplasmic reticulum and failure of amino acid incorporation into liver proteins. Hepatic necrosis [10], [15] and apoptosis [16] result.

Thioacetamide causes hepatic failure in rats using protocols of varying dose and timing [10], [17], [18]. Reproducibility of the model is affected by several factors, such as intra-species variation, age, diet, and also concomitant supportive therapy. We characterized the clinical, biochemical, metabolic and histological pattern of thioacetamide-induced hepatic failure in the Wistar rat, to create a model of substantial severity (∼70% mortality), which fulfils the criteria of reproducibility, reversibility and provision of a therapeutic window, suggested by Terblanche et al. [19] for animal models of AHF.

Previous reports illustrate contradictory findings following NO inhibition in experimental AHF, in part reflecting choice of iNOS inhibitor, doses of hepatotoxin and precise experimental design [6], [10], [20]. We report here the effect of two inhibitors of NO synthesis, l-NAME (NG-nitro-l-arginine methyl ester), a relatively non-specific inhibitor of NO synthesis, and AMG, a more selective inhibitor of iNOS [21], on the course of thioacetamide-induced AHF in rats. We also investigated the effect of preloading animals with l-arginine, which by providing the substrate for iNOS should exaggerate the consequences of NO generation. Our results indicate that NO is involved in the pathogenesis of thioacetamide induced liver damage and AHF in rats, and selective iNOS inhibition minimizes the severity of TAA-induced damage induced subsequently.

Section snippets

Animals and treatments

Male Wistar rats weighing 250–300 g were obtained from Charles River (UK). The rats were fed with standard rat chow diet ad libitum, allowed free access to water and housed in standard facilities with a 12:12-h day/night cycle at a room temperature of 25 °C. All chemicals were reagent grade and were obtained from Sigma Chemical Co. (St. Louis, MO).

Seventy animals (n=10 per Group) were allocated to seven different regimes (Groups I–VII) to monitor encephalopathy and to assess survival over 96 h

Results

Preliminary studies explored the effects of 100–600 mg/kg body weight TAA dissolved in 0.9% saline given as single or repeated intraperitoneal injections, and assessed the effects of food withdrawal for up to 12 h (to reduce intracellular glutathione stores and diminish intracellular free radical scavenging [23]). Two injections of TAA 500 mg/kg body weight given 8 h apart produced a model with clinical, biochemical and histological similarities of AHF in man, including encephalopathy and

Discussion

Thioacetamide induces hepatocyte damage following its metabolism to thioacetamide sulphene and sulphone, via a critical pathway involving CYP4502E1-mediated biotransformation [13], [24]. We examined the role of NO in the pathophysiology of this model by use of regimes designed both to exacerbate and to limit NO production, with the particular aim of defining approaches that could ameliorate the severity of hepatic failure. We therefore utilized a model of liver failure that induces severe

Acknowledgements

Tony Rahman held a Dunhill Clinical Fellowship. The work was supported by the Liver Group Charity.

References (26)

  • C.R. Gardner et al.

    Role of nitric oxide in acetaminophen-induced hepatotoxicity in the rat

    Hepatology

    (1998)
  • P.Y. Martin et al.

    Nitric oxide as a mediator of hemodynamic abnormalities and sodium and water retention in cirrhosis

    N Engl J Med

    (1998)
  • A. Ellis et al.

    Circulatory, respiratory, cerebral, and renal derangements in acute liver failure: pathophysiology and management

    Semin Liver Dis

    (1996)
  • Cited by (45)

    • Potential Effect of 1,25 Dihydroxyvitamin D<inf>3</inf> on Thioacetamide-Induced Hepatotoxicity in Rats

      2019, Journal of Surgical Research
      Citation Excerpt :

      Previous studies have shown the role of iNOS in the pathophysiological mechanisms that lead to postnecrotic regeneration after TAA administration.29 This observation is in the same parallel with the findings of a study reporting the reduction of TAA-induced hepatotoxicity and the improvement of mortality by aminoguanidine, a selective iNOS inhibitor, but not by nonselective NOS inhibitor.30 In the present study, enhanced iNOS expression by TAA administration and its suppression by 1,25(OH)2D3 might be associated with the inhibition of NF-κB activation.

    • Kinetic deterioration of short memory in rat with acute hepatic encephalopathy: Involvement of astroglial and neuronal dysfunctions

      2019, Behavioural Brain Research
      Citation Excerpt :

      At the histological level, our TAA rats exhibited a kinetic deterioration of the liver tissue with inflammation and hepatocyte necrosis without the presence of features of liver fibrosis. The liver tissue responses to acute TAA treatment is well documented [34,35], as previously mentioned, acute TAA toxicity leads to the prominent changes in the liver tissue architecture with appearance of necrotic hepatocytes and inflammatory cells, mostly macrophages, around the central vein [36]. Meanwhile, ALF triggered an obvious progressive decline of short memory performance visualized by the loss of alternation behavior at 12, 24 and 36 h stages of AHE.

    • The protective effect of propofol against TNF-α-induced apoptosis was mediated via inhibiting iNOS/NO production and maintaining intracellular Ca<sup>2+</sup> homeostasis in mouse hippocampal HT22 cells

      2017, Biomedicine and Pharmacotherapy
      Citation Excerpt :

      NO is produced from NOS, which has three isoforms including endothelial nitric oxide synthase (eNOS), neuron nitric oxide synthase (nNOS) and inducible nitric oxide synthase (iNOS). In central nervous system, iNOS serve as the main source of NO upon exposure to pro-inflammatory cytokine [32]. NO may induce cytotoxicity through several mechanisms.

    View all citing articles on Scopus
    View full text