Differential expression of stress-inducible proteins in chronic hepatic iron overload

https://doi.org/10.1016/j.taap.2007.05.011Get rights and content

Abstract

Introduction:

Oxidative stress can trigger a cellular stress response characterized by induction of antioxidants, acute phase reactants (APRs) and heat shock proteins (HSPs), which are presumed to play a role in limiting tissue damage. In rodents, hepatic iron overload causes oxidative stress that results in upregulation of antioxidant defenses with minimal progressive liver injury. The aim of this study was to determine whether iron overload modulates expression of other stress-responsive proteins such as APRs and HSPs that may confer protection against iron-induced damage in rodent liver.

Methods:

Male rats received repeated injections of iron dextran or dextran alone over a 6-month period. Hepatic transcript levels for a panel of APRs and HSPs were quantitated by real-time PCR and protein expression was evaluated by Western blot and immunohistochemistry.

Results:

Hepatic iron concentrations were increased > 50-fold in the iron-loaded rats compared to controls. Iron loading resulted in striking increases in mRNAs for Hsp32 (heme oxygenase-1; 12-fold increase vs. controls) and metallothionein-1 and -2 (both increased ∼ 6-fold). Transcripts for α1-acid glycoprotein, the major rat APR, were increased ∼ 3-fold, while expression of other classical APRs was unaltered. Surprisingly, although mRNA levels for the HSPs were not altered by iron, the abundance of Hsp25, Hsp70 and Hsp90 proteins was uniformly reduced in the iron-loaded livers, as were levels of NAD(P)H:quinone oxidoreductase 1, an Hsp70 client protein.

Conclusions:

Chronic iron administration elicits a unique pattern of stress protein expression. These alterations may modulate hepatic responses to iron overload, as well as other injury processes.

Introduction

Hepatic iron overload resulting from hemochromatosis or hematologic disorders such as thallasemia can cause cirrhosis, end-stage liver disease and hepatocellular carcinoma. Iron-catalyzed oxidative damage to cellular components is presumed to be the major causal factor in the development of hepatic fibrogenesis and carcinogenesis (Kowdley, 2004). In support of this concept, removal of excess iron by phlebotomy or chelation prevents the development of these complications. Interestingly, however, recent data indicate that hepatic complications of iron overload in individuals with hemochromatosis are much less common than previously assumed (Beutler et al., 2002). Furthermore, although significant hepatic iron burdens can be produced in experimental animals by a variety of methods, these models rarely result in significant liver fibrosis (Ramm, 2000), which is the precursor lesion of serious liver pathology in human iron overload. Taken together, these observations suggest that mammalian livers possess a substantial capacity to resist the toxicity of chronic iron exposure.

The mechanisms that confer resistance to iron-induced hepatotoxicity are incompletely understood. We and others have reported that iron administration is associated with enhanced glutathione synthetic machinery in rodent liver (Ogino et al., 1989, Brown et al., 1998, Brown et al., 2007). In addition, enzymes involved in glutathione-dependent detoxification of reactive intermediates are upregulated by iron loading (Kahn et al., 1995). Given that excess iron is presumed to increase oxidant production, upregulation of antioxidant defenses is likely an important factor contributing to resistance to iron-related toxicity. However, whether other types of protective mechanisms are induced in response to iron overload has not been investigated.

In other models of tissue injury such as those resulting from thermal injury, radiation or heavy metal administration, induction of antioxidant expression frequently occurs in parallel with the upregulation of other stress-inducible genes whose products are broadly regarded as playing a role in limiting tissue damage. These include the acute phase reactants (APRs) and the heat shock proteins (HSPs). The acute phase reaction is characterized by altered transcription (positive or negative) of numerous target genes, many of which are expressed exclusively or predominantly by the liver (Gabay and Kushner, 1999). The products of these genes (APRs) participate in diverse processes such as blood coagulation, innate immunity, sequestration of metals, antiproteolytic activities, etc. The acute phase reaction is typically triggered by localized or systemic inflammation, and cytokines such as IL-1 and IL-6 are important mediators of this response.

Although it is clear that inflammatory mediators are the primary stimulus for the acute phase reaction, oxidative stress appears to play a role in this response. For example, the acute phase reaction caused by localized inflammation is accompanied by evidence of hepatic oxidative stress (Proulx and du Souich, 1995, El-Kadi et al., 2000), while treatments that deplete hepatic glutathione stores increase the expression of some APRs (Tacchini et al., 2002). Notably, hepcidin and ferritin, two proteins that play key roles in the regulation of iron metabolism and whose expression is induced by iron, are also APRs (Nemeth et al., 2003, Pigeon et al., 2001). These observations suggest that iron-induced oxidative stress may stimulate expression of other APRs that contribute to protection from liver injury.

The HSPs are another important class of proteins whose expression is inducible by a variety of stressors. Collectively, the HSPs are believed to assist in the proper folding of newly synthesized proteins and/or in the refolding of denatured proteins. Destabilization and unfolding of proteins leading to aggregation have been proposed to be the common signal that elicits the heat shock response (Freeman et al., 1999). Although destabilization and unfolding occur in response to elevated temperature, oxidation of thiol or other residues can have similar effects on protein stability, thus accounting for the observation that treatments that cause oxidative stress frequently induce expression of heat shock proteins as well. In support of the relationship between oxidative stress and increased HSP expression, we have previously reported that Hsp32 (heme oxygenase-1, HO-1) levels are increased in iron-loaded livers (Brown et al., 2003). However, it is not known whether other HSPs are altered by iron loading. Therefore, the goal of these studies was to determine the effects of chronic iron overload on the expression of a variety of stress-inducible proteins including APRs and HSPs.

Section snippets

Methods

Male Sprague–Dawley rats weighing 200–250 g (Harlan Laboratories, Indianapolis, IN) were individually housed in polyethylene cages with stainless steel tops and were fed a standard rat diet (Dyets, Inc., Bethlehem, PA) and allowed water ad libitum. Iron dextran (50 mg iron per rat for the first 4 injections, 100 mg per rat thereafter) was administered by IP injection every 2 weeks for 6 months as previously described (Brown et al., 2007). Control animals received IP injections of an equivalent

Hepatic iron concentrations, histology and hydroxyproline content

Median hepatic iron concentrations were increased > 50-fold in the iron-loaded rats (10,706 μg/g liver) compared to controls (189 μg/g; p < 0.001). Histochemical staining for storage iron demonstrated no stainable iron in control livers; large sinusoidal iron deposits (siderotic nodules) and heavy hepatocellular iron deposition were seen in the rats treated with iron dextran (Figs. 1A, B). In line with our previous observations (Brown et al., 1998, Brown et al., 2003), there was a modest but

Discussion

Iron overload resulting from hemochromatosis or hematologic disease can cause progressive hepatic fibrosis and cirrhosis in humans. In contrast, as this work demonstrates, iron overload is a weak fibrogenic stimulus in rodent liver. Likewise, while increased levels of hepatic iron in human liver are commonly believed to accelerate liver fibrosis in the presence of chronic viral hepatitis or alcoholic liver disease (Alla and Bonkovsky, 2005), the combination of iron with other hepatotoxins does

Acknowledgments

KEB was supported by a Merit Review grant from the Veterans Administration. The authors thank Dr. Joe Cullen for the gift of the NQOI antibody.

References (40)

  • E. Nemeth et al.

    Hepcidin, a putative mediator of anemia of inflammation, is a type II acute-phase protein

    Blood

    (2003)
  • T. Ogino et al.

    Stimulation of glutathione synthesis in iron-loaded mice

    Biochim. Biophys. Acta

    (1989)
  • J.R. Oliver et al.

    Augmented hepatic injury followed by impaired regeneration in metallothionein-I/II knockout mice after treatment with thioacetamide

    Toxicol. Appl. Pharmacol.

    (2006)
  • J. Olynyk et al.

    A long-term study of the interaction between iron and alcohol in an animal model of iron overload

    J. Hepatol.

    (1995)
  • C. Pigeon et al.

    A new mouse liver-specific gene, encoding a protein homologous to human antimicrobial peptide hepcidin, is overexpressed during iron overload

    J. Biol. Chem.

    (2001)
  • X. Préville et al.

    Mammalian small stress proteins protect against oxidative stress through their ability to increase glucose-6-phosphate dehydrogenase activity and by maintaining optimal cellular detoxifying machinery

    Exp. Cell Res.

    (1999)
  • L. Tacchini et al.

    Activation of transcription factors by drugs inducing oxidative stress in rat liver

    Biochem. Pharmacol.

    (2002)
  • P.J. Thornalley et al.

    Possible role for metallothionein in protection against radiation-induced oxidative stress. Kinetics and mechanisms of its reaction with superoxide and hydroxyl radicals

    Biochim. Biophys. Acta

    (1985)
  • G.S. Wang et al.

    Dietary iron overload inhibits carbon tetrachloride-induced promotion in chemical hepatocarcinogenesis: effects on cell proliferation, apoptosis, and antioxidation

    J. Hepatol.

    (1999)
  • V. Alla et al.

    Iron in nonhemochromatotic liver disorders

    Semin. Liver Dis.

    (2005)
  • Cited by (24)

    • Hepcidin and Iron Metabolism in Experimental Liver Injury

      2021, American Journal of Pathology
      Citation Excerpt :

      A small peptide produced by the liver, hepcidin causes degradation of the iron export protein ferroportin, which is required for the transfer of iron from enterocytes and macrophages to the systemic circulation.7 Numerous investigators using a variety of methods of iron administration have demonstrated increases in Hamp mRNA in response to iron.5,8,9 These results support the prevailing paradigm whereby hepcidin expression is upregulated by increased iron stores.

    • Altered expression of iron regulatory proteins with aging is associated with transient hepatic iron accumulation after environmental heat stress

      2014, Blood Cells, Molecules, and Diseases
      Citation Excerpt :

      Ferroportin is the receptor for hepcidin, and it is degraded intracellularly after hepcidin binding [20]. We have previously reported that hepcidin mRNA expression is increased 2.5-fold in the cohort of chronically iron-loaded rats used for comparison in this study [30]; however, hepcidin expression was not altered with aging (Bloomer, Kregel, Brown, manuscript submitted). Thus, in the chronically iron-loaded animals, the reduction in levels of Fpn protein is likely attributable to the upregulation in hepcidin.

    • Heat stress stimulates hepcidin mRNA expression and C/EBPα protein expression in aged rodent liver

      2014, Archives of Gerontology and Geriatrics
      Citation Excerpt :

      Each sample was also assayed for 18S expression using TaqMan 18S ribosomal RNA control reagents (Applied Biosystems, Foster City, CA) and expression of the gene of interest was normalized to 18S expression. To quantitate target gene mRNA expression, the comparative Ct method was utilized and results are expressed as 2−ΔΔCt as described previously (Brown, Broadhurst, Mathahs, & Weydert, 2007). Nonheated animals in each age group served as the reference control and samples were run in triplicate on each plate.

    • Concomitant apoptosis and regeneration of liver cells as a mechanism of liver-tumor promotion by β-naphthoflavone involving TNFα-signaling due to oxidative cellular stress in rats

      2011, Toxicology
      Citation Excerpt :

      An increase in GST-P+ single cells has been observed with Cu-overloading without DEN-initiation, which was suggested to be cytoprotective against accumulating Cu (Abe et al., 2008). Similarly, HO-1 has been shown to protect against oxidative stress in liver cells (Nakahira et al., 2003; Brown et al., 2007). Recently, we observed a profound increase in single liver cells positive for GST-P or HO-1 as well as elevated TBARS levels in Cu-overloaded animals, in parallel with the tumor-promoting activity of Cu, in the same two-stage hepatocarcinogenesis model as used here (Mizukami et al., 2010).

    • Tumor promotion by copper-overloading and its enhancement by excess iron accumulation involving oxidative stress responses in the early stage of a rat two-stage hepatocarcinogenesis model

      2010, Chemico-Biological Interactions
      Citation Excerpt :

      Experimentally, Cu-overloading induces MTs in the liver [33]. Although MTs do not bind to Fe [34], Fe overloading has shown to increase MT-1/2-expression in liver cells [24]. However, in the two-stage hepatocarcinogenesis, we could not find any association of MT-1/2+ foci with GST-P-immunoreactivity or cell proliferation activity by Cu- and/or Fe-overloading in the present study, in accordance with our previous results showing no association with tumor-promoting activity [17].

    View all citing articles on Scopus
    View full text