metricas
covid
Buscar en
Annals of Hepatology
Toda la web
Inicio Annals of Hepatology Oxidative stress: A radical way to stop making bile
Journal Information
Vol. 7. Issue 1.
Pages 16-33 (January - March 2008)
Share
Share
Download PDF
More article options
Visits
3833
Vol. 7. Issue 1.
Pages 16-33 (January - March 2008)
Open Access
Oxidative stress: A radical way to stop making bile
Visits
3833
Marcelo G. Roma1,
Corresponding author
roma@ifise.gov.ar

Address for correspondence:
, Enrique J. Sanchez Pozzi1
1 Instituto de Fisiología Experimental (IFISE) - Facultad de Ciencias Bioquímicas y Farmacéuticas (CONICET - U.N.R.), S2002LRL, Rosario, ARGENTINA
This item has received

Under a Creative Commons license
Article information
Abstract
Full Text
Bibliography
Download PDF
Statistics
Figures (4)
Show moreShow less
Abstract

Oxidative stress is a common feature in most hepatopathies. In recent years, evidence has accumulated that reactive oxygen species (ROS) induce a number of functional changes either deleterious or adaptive in the capability of the hepatocytes to produce bile and to secrete exogenous and endogenous compounds. This review is aimed to describe the mechanisms involved in these alterations. For this purpose, we will summarize:

  • 1)

    The current evidence that acutely-induced oxidative stress is cholestatic, by describing the mechanisms underlying the hepatocyte secretory failure, including the disorganization of the actin cytoskeleton and its most noticeable consequences, the impairment of tight-junctional structures and the endocytic internalization of canalicular transporters relevant to bile formation.

  • 2)

    The role for oxidative-stress-activated signalling pathways in the pathomechanisms described above, particularly those involving Ca2+ elevation and its consequent activation of Ca2+-dependent PKC isoforms.

  • 3)

    The mechanisms involved in the adaptive response against oxidative stress mediated by ROS-responsive transcription factors, involving up-regulation of GSH-synthesizing enzymes, GSH-detoxifying enzymes and the hepatocellular efflux pumps; this response enhances the co-coordinated inactivation by GSH conjugation of lipid peroxides and their further cellular extrusion.

  • 4)

    The manner this adaptive response can be surpassed by the sustained production of ROS, thus inducing transcriptional and posttranscriptional changes in transporters relevant to bile formation, as has been shown to occur, for example, after long-term administration of aluminum to rats, in the Long-Evans Cinnamon rat (a model of chronic hepatic copper accumulation mimicking Wilson’s disease), and in ischemia-reperfusion injury.

Key words:
Bile secretion
oxidative stress
calcium
signalling
cytoskeleton
transcription factor
canalicular transporters
Full Text
1. Introduction

Reactive oxygen species (ROS) are formed during a variety of biochemical reactions and cellular functions. For example, leakage of electrons leading to superoxide anion (O2•—) generation may occur during the electron transport through the mitochondrial respiratory chain.1 Similarly, O2•— may be generated during the microsomal, cytochrome-P450-mediated hydroxylation of endo and exogenous compounds for detoxification. This is due to uncoupling of electrons during their transfer from NADPH cytochrome P450 reductase to cytochrome P450, or from cytochrome P450 to its substrates,2 as was shown to occur for ethanol3 and benzo(a)pyrene4 metabolisms, among many others. This steady-state ROS formation is balanced by a similar rate of consumption by the antioxidant defences provided by enzymes (e.g., superoxide dismutase, catalase, glutathione peroxidase, glutathione-5-transferase [GST]) and free-radical-scavenging endogenous and exogenous compounds (e.g., glutathione, ascorbate, α-tocopherol, β-carotene).5

Oxidative stress results from an imbalance between endogenous pro-oxidant formation and neutralization by antioxidant defences. When this occurs, ROS can damage virtually all types of biological molecules. These molecular alterations trigger a number of interrelated pathomechanisms, namely: i) free-thiol oxidation of proteins;6,7ii) disassembly of cytoskeleton,8,9 with plasma membrane blebbing as a major emergent;10iii) formation of mitochondrial permeability transition pores, leading to the release of mitocondrial promoters of apoptosis (cytochrome c, Smac/DIABLO) and uncoupling of the electron transport chain from ATP production, with eventual necrosis associated with ATP depletion;11iv) increased plasma-membrane peroxidation, leading to membrane permeabilization and leakage of cytosolic components;12v) oxidative breaking of DNA strands, leading to genotoxicity and carcinogenesis;13,14vi) elevation of intracellular Ca2+ levels, which facilitates mitochondrial permeability transition and activation of Ca2+-dependent proteases, endonucleases, and lipases.11,15

Due to its multiple energy-dependent functions, liver has a high mitochondrial metabolic rate, and is heavily engaged in detoxification mechanisms that involve redox-enzyme systems. Since these are major sources of endogenous free radicals, ROS production is higher in liver as compared with most organs. Hence, hepatocytes are equipped with abundant antioxidant enzymes to counterbalance this oxidative challenge.16 However, this borderline equilibrium makes liver highly susceptible to the pro-oxidant injury induced by pathological conditions that disrupt this balance, by enhancing ROS formation, by impairing the antioxidant defences, or both.17-19 Indeed, oxidative stress is a common feature in most hepatopathies including hepatic ischemia-reperfusion injury following hepatectomy or liver transplantation,20,21 obstructive cholestasis,22 chronic cholestatic liver diseases,23-27 sepsis-induced cholestasis,28 viral,29,30 toxic31 and autoimmune32 hepatitis, alcoholic30,33 and non-alcoholic34-36 steatohepatitis, and pathologies leading to hepatic accumulation of metals, such as iron (hemochromatosis, iron-loading anaemia)37,38 or copper (Wilson’s disease).39

Much attention has been focused on the mechanism of oxidative-stress-induced hepatocellular death.40-43 However, this usually involves only a limited number of hepatocytes. For example, in patients with hepatic oxidative stress due to chronic alcoholic intake, ~7 per 1000 hepatocytes suffer apoptosis,44 and only marginal necrosis has been observed in the liver of patients with a similar alcohol intake.45 The question therefore arises as to which extent the bulk of the remaining viable cells is functionally affected by the oxidative injury.

In recent years, evidence has accumulated that oxidative stress is cholestatic. Functional changes involve impairment of hepatocellular biliary secretory machinery through both the direct oxidative damage of relevant cellular structures by ROS or, indirectly, by modification of intracellular signal transduction pathways sensitive to changes in the intracellular redox state. This review is aimed to summarize the mechanisms involved in these alterations.

2. Mechanisms of hepatobiliary secretory function: «Let it bile»

Bile formation is an osmotic process driven by the vectorial transport of certain solutes into bile. They comprise mainly bile salts and both oxidized (GSSG) and reduced (GSH) glutathione. For these solutes to induce blood-to-bile water transport primarily, they need to be concentrated and retained into a confined space (the bile canaliculus). The hermeticity is provided by tight-junctional structures localized in the paracellular pathway, near the apical surface of the cells.

Once secreted, these solutes induce passive water movement in response to osmotic gradients via both paracellular and transcellular routes. Transcellular water movements are facilitated by the water channels, aquaporins (AQP) type 9 and 8, located at the basolateral and apical membranes, respectively.46

As expected for a solute exerting osmotic forces at the canalicular level, a linear relationship between bile salt secretion and bile flow has been observed in all vertebrates, including man. This bile flow fraction is conventionally referred to as «bile salt-dependent fraction of the bile flow».47 Since a positive value is, in most cases, obtained when extrapolating bile-salt output to zero, a bile salt-independent fraction of the bile flow is also apparent, which has been mainly attributed to glutathione output. This primary (canalicular) secretion is further modified by cholangiocytes during its transit along bile ducts, as a result of a balance between hormone-dependent water and electrolyte secretion and, on the other hand, the obligatory absorption of water, electrolytes and organic solutes.48,49Figure 1 depicts the main solutes and transporters relevant to bile formation at both the hepatocellular and the bile-ductular levels.

Figure 1.

Main transport proteins and metabolic events involved in bile-flow generation and in the hepatic handling of the endogenous organic anion, bilirubin (Br) and of the model exogenous cholephilic organic anion, sulphobromophthalein (BSP). Primary, ATP-dependent transporters are depicted in dark grey. For the sake of simplicity, only rodent transporters are shown, but human orthologs have been identified for all of them (see section 2 for details). Na+-dependent uptake of bile salts (BSs) at the sinusoidal level is mediated by the Na+-taurocholate cotransporting polypeptide (Ntcp), driven by the Na+ gradient generated by Na+/K+-ATPase. BSs are also taken up in a Na+-independent manner by the organic anion-transporting polypeptide (Oatp) family of transporters, which transport a wide range of solutes, including endogenous and exogenous organic anions, such as unconjugated Br and BSP. After traversing the cell bound to cytosolic protein, taurine or glycine-conjugated, monovalent BSs are excreted at the canalicular pole by the bile salt export pump (Bsep). Sulfated or glucuronidated, divalent BSs are instead transported by the multidrug resistance-associated protein 2 (Mrp2). Following their Oatp-mediated uptake, Br is conjugated with uridine diphosphate (UDP)-glucuronic acid (UDPGA) by the endoplasmic-reticulum enzyme, UDP-glucuronosyltransferase (UDPGT), to form Br mono/di-glucuronides (Br-gluc), which are excreted by Mrp2. BSP is conjugated with reduced glutathione (GSH) by the cytosolic enzyme, glutathione 5-transferase, to form the glutathione-conjugated parent compound BSP-GSH, which can be also excreted by Mrp2. GSH available for biliary excretion is provided by de novo synthesis. GSH biosynthesis comprises a two-step reaction catalyzed by the enzymes, γ-glutamyl-cysteinyl synthetase (γ-GCS) and GSH synthetase (GS). γ-GCS catalyzes the formation of L-γ-glutamyl-cysteine (γ- GC) from L-glutamate (Glu) and L-cysteine (Cys), whereas GS catalyzes GSH synthesis by further incorporation of L-glycine (Gly). GSH is excreted with high affinity by an as yet unidentified high-affinity canalicular GSH transporter, whereas its oxidized form, GSSG, is excreted with high affinity by Mrp2. Some cross-transport may however exist, particularly when any of these transporters is saturated (not shown). Both BSs and GSH/GSSG can act as primary driven force for osmotic bile formation, by promoting water movements via AQP9 and AQP8, localized at the sinusoidal and canalicular membrane domains, respectively. Canalicular bile flow is further modified during its transit along the bile ducts by both secretory and absorptive processes. Ductular fluid secretion is driven by the secretin-regulated, cAMP-dependent output of a HCO3--rich fluid facilitated by the anion exchanger 2 (Ae2). Exchange is driven by the out-to-in Cl--concentration gradient created by the ATP-dependent, secretin-activated channel, cystic fibrosis transmembrane regulator (CFTR). Blood-to-bile water movement is facilitated by constitutive AQP4 in the basolateral membrane, and secretin-stimulated AQP1 in the apical membrane. Absorption is driven by the osmotic gradients created by Glu (through unidentified transporters), glucose (Glc; transported by SGLT1 and GLUT1 at the apical and basolateral domains, respectively) and BSs (taken up by the apical Na+-dependent bile salt transporter, Asbt, followed by basolateral extrusion via the multidrug resistance-associated protein 3, Mrp3, and the heterodimeric organic solute transporter, Ostα-Osβ).

(0.22MB).

Bile salts are the predominant organic solutes in bile. The main sinusoidal transport system for bile-salt uptake is the Na+-taurocholate cotransporting polypeptide, which has been cloned from both rat (Ntcp, Slc10a1)50 and human liver (NTCP, SLC10A1).51 Ntcp/NTCP is driven by a transmembrane Na+ gradient maintained by the Na+/K+-ATPase pump, which is also strategically localized in the sinusoidal membrane.52 Ntcp/NTCP accounts for the transport of more than 80% of amidated bile salts (the major circulating bile salts), and only 40% of their unconjugated, parent compounds.53 The remaining fraction of circulating bile salts is taken up by a non-electrogenic, Na+-independent transport system, formed by a family of transporters collectively named organic aniontransporting polypeptides (Oatps/OATPs for rat and human, respectively).54 Humans possess four OATPs: OATP1A2 (SLCO1A2/SLC21A3), OATP1B1 (SLC21A6), OATP1B3 (SLC21A8) and OATP2B1 (SLC21A9). In the rat, there are three ones: Oatp1a1 (Slc21a1), Oatp1a4 (Slc21a5) and Oatp1b2 (Slc21a10). Apart from bile salts, Oatps/OATPs accept a wide range of amphipathic, organic compounds, including bilirubin, bilirubin glucuronides, leukotrienes, estrogens, «type II» organic cations and several exogenous organic anions, with the cholephilic dye sulphobromophthalein (BSP) being a prototypical example of the latter ones.55 Certain specificity exists among Oatp isoforms. All but OATP2B1 are involved in bile salt uptake in humans,56 whereas bilirubin diglucuronides and, perhaps, unconjugated bilirubin are transported only by OATP1B1.57 In rats, all of the iso forms are involved in uptake of conjugated and unconjugated bile salts,56 whereas only Oatplal and Oatp1a4 are involved in bilirubin monoglucuronide transport.58

After traversing the cell by Fick’s diffusion bound to high-affinity cytosolic proteins, monoanionic bile salts (C24 amides conjugated with glycine or taurine) are excreted in the canalicular pole by the bile salt export pump (BSEP/Bsep), an ATP-binding cassette transporter.59 In contrast, canalicular efflux of divalent, bipolar sulphated or glucuronidated bile salts is mediated by the multidrug resistance-associated protein 2 (MRP2/Mrp2; ABCC2/Abcc2). This carrier is also engaged in the biliary excretion of endogenous and exogenous non-bileacid organic anions conjugated with glutathione, glucuronic acid or sulphate. They comprise leukotriene C4, bilirubin glucuronides, some steroid sulphates and cholephilic dyes, including BSP both in its unconjugated and glutathione-conjugated forms.60

The bile-salt-independent fraction of the bile flow depends on glutathione excretion, mainly in its reduced form, GSH (~80%).61,62 Hepatocellular glutathione transport mechanisms are poorly understood. The liver is the main site of glutathione synthesis, exporting this peptide into both blood and bile. Most, if not all biliary glutathione comes from this intracellular source.63 No canalicular transporter for glutathione has been cloned so far. However, a high-affinity, electrogenic carrier has been functionally characterized.64-67 This transporter exports actively GSH into bile, and can transfer GSSG and GSH conjugates as well, but with lower affinity. Another transporter likely involved in glutathione canalicular transport is Mrp2. However, this carrier bears low affinity towards GSH, although it can transfers GSSG and GSH-conjugates with high affinity.65

Canalicular bile flow is further modified during its transit along bile ducts by both secretory and absorptive processes.68 Ductular fluid secretion is mainly driven by the secretin-regulated, cAMP-dependent output of a HCO3--rich fluid facilitated by the Cl-/HCO3--exchange system, anion exchanger 2 (Ae2/AE2). Exchange is dependent on the out-to-in Cl-concentration gradient, which is maintained by the Cl- efflux across the apical membrane via the ATP-dependent, secretin-activated, cystic fibrosis transmembrane regulator (CFTR). Blood-to-bile water movement at the ductular level is facilitated by constitutive AQP4 in the basolateral membrane, and secretin-stimulated AQP1 in the apical membrane.46 On the other hand, absorption of ductular water and electrolytes is driven by the osmotic gradients created by bile-to-plasma transport of electrolytes and organic solutes. They comprise: i) glutamate, transported by as yet unidentified carriers; ii) glucose, transported by SGLT1 and GLUT1 at the apical and basolateral domains, respectively; iii) bile salts, taken up by the apical Na+-dependent bile salt transporter, ASBT/Asbt (SLC10A2/slc10a2), and extruded by both the basolateral export pump, MRP3/Mrp3 (ABCC3/Abcc3), and the heterodimeric organic solute transporter, OSTα-OSTβ/Ostα-Ostβ.49,69

3. Changes in hepatobiliary secretory function induced by oxidative stress

Compelling evidence in the literature indicates that oxidative challenge affects the hepatocyte-secretory machinery by impairing both bile flow (hepatocellular cholestasis) and the biliary excretion of endo and xenobiotics.

Oxidative-stress-induced impairment of bile flow generation has been demonstrated to occur after exposure to a number of pro-oxidant agents, including. tert-butylhydroperoxide (tBOOH),70,71 hydrogen peroxide,70,72 menadione,73 allyl alcohol,73 ethylhexanol,73 chloro-dinitrobenzene,71 CCl4,74 ethacrynic acid75 and lindane,76 among others. Some pharmacological agents, such as cyclosporine A,77 dapsone78,79 and nitrofuran derivatives,80,81 also induce cholestasis due, at least in part, to their prooxidant properties. Finally, maneuvers leading to hepatic oxidative stress, such as hepatic82,83-87 and intestinal88 ischemia-reperfusion, or aluminum intoxication,89,90 also induce bile flow impairment.

These cholestatic conditions are often associated with impairment of the bile salt-dependent bile flow. In many of these cases, the decrease in the overall bile flow was apparent even when the bile salt-independent bile flow was enhanced due to augmented GSSG output; GSSG extrusion into bile helps to maintain the intracellular GSH/GSSG ratio constant, and hence the intracellular redox equilibrium.70,91 However, under milder oxidative stress conditions, total bile flow may not change. In this case, the decrease in the bile salt-dependent bile flow induced by ROS may not be enough to counteract the parallel increase in the bile salt-independent bile flow due to enhanced GSSG biliary excretion.70,92 Similarly, osmotic choleresis associated with the biliary excretion of metabolites of the pro-oxidant agents used may mask ROS-induced cholestasis, as occurs soon after the administration of ethacrynic acid.70,75,93 However, we can consider these situations as cholestatic as well, since at least part of the hepatocyte machinery involved in bile formation is impaired.

3.1. Impairment of the hepatobiliary function by oxidative stress: «The orthodox view»

The classical view to interpret the cholestasis associated to pro-oxidant conditions is based upon the occurrence of the following pathomechanisms:

  • i)

    Oxidative-stress-induced hepatocellular cell death by necrosis41 and apoptosis,42,94 which produces a reduction of the number of living, functional parenchymal cells. This pathomechanism is mainly associated with Ca2+-facilitated mitochondrial permeability transition, followed by release of mitochondrial pro-apoptotic factors and impairment of the electron transport chain, leading eventually to ATP depletion and necrotic cell death.95•—96

  • ii)

    Impairment of the bile salt-dependent fraction of the bile flow due to competitive inhibition of bile salt transport by the intracellular GSSG formed in excess during the oxidative challenge.70,72 This is supported by the fact that GSSG cis-inhibits the transport of the model bile salt, taurocholate, in liver canalicular membrane vesicles.97 In line with this contention, mirror curves depicting an increase in biliary excretion of GSSG and a decrease in biliary excretion of bile salts were obtained by administrating different pro-oxidant compounds in the isolated rat perfused liver, such as hydrogen peroxide,72 menadione92 and tBOOH.70,72 Furthermore, an inverse relationship between intracellular GSSG content and the percentage of inhibition of the biliary bile salt output was obtained by manipulating GSSG content using different oxidizing compounds, in the presence or absence of inhibitors of GSSG formation.72,92

  • iii)

    Impairment of the bile salt-independent bile flow due to a decrease in the biliary excretion of total glutathione (GSH plus GSSG). This occurs as a consequence of a depletion of the hepatic levels of glutathione due to the sustained plasmatic and biliary exportation from the cell as GSSG to maintain the GSH/ GSSG ratio.83

3.2. Oxidative stress and hepatobiliary function: «The Actin Connection»

The above-mentioned mechanisms may be predominant under strong oxidizing conditions. However, it has become increasingly apparent that under mild, or even low, oxidizing conditions, functional alterations in the hepatocyte capability to produce bile may occur. Indeed, oxidative stress-induced drop of bile flow and/or decrease of the bile salt-secretory rate becomes aparent before leakage of cytosolic hepatocellular enzymes or increments in intracellular GSSG levels is induced by administration of different pro-oxidant compounds to isolated perfused rat livers.70,75 Likewise, in isolated rat hepatocyte couplets, an in vitro model for the study of hepatocanalicular function, the secretion of fluorescent bile-salt analogues into the canalicular vacuole was impaired by low concentrations of the pro-oxidant compounds, tBOOH and 2,3-dimethoxy-1,4-naphthoquinone, even when GSSG intracellular levels and cellular viability were unaffected.98 Finally, also in the couplet model, a dissociation between apical secretion of fluorescent bile-salt analogues and the amount of GSSG produced in the cell by the oxidizing effects of redox-cycling quinones has been reported.99 Overall, these results suggest that more subtle changes in the machinery involved in bile formation occur under mild oxidative stress conditions. The actin cytoskeletal disruption, which occurs even at very low oxidative stress levels, seems to be a crucial, causal factor.

The actin cytoskeleton is a dynamic network of filamentous actin (F-actin), formed by reversible assembly of monomeric actin (G-actin). It plays a crucial role in cell motility and in cell-shape changes occurring during the cellular cycle.100 In addition, actin is essential for the control of cell-cell interactions.100 For example, F-actin is anchored to Zonula occludens-associated proteins, such as ZO-1, ZO-2, ZO-3, 7H6 and cingulin, thus regulating the paracellular permeability by changes in its contractility status/organization.101 Actin is also involved in transcitosis processes, by operating as a ‘bridge’ between microtubules and the apical membrane itself, in a coordinated action of the microtubule-and the F-actin-based motor proteins, kinesin and myosin, respectively.102 Actin can also interact with, and possibly regulate, transmembrane proteins via binding to interacting-partner proteins, such as PDZ and HAX-1. These cytoskeleton-associated proteins are required for the biosynthetic targeting of transmembrane proteins from the trans-Golgi network to the proper membrane domain, and for their further cell-surface retention.103-105

The actin cytoskeleton is one of the primary targets of oxidative stress.8 The oxidative challenge promotes the oxidation of actin at a sulfhydryl group of cysteine in position 374.8 This induces conspicuous changes in actinspatial distribution, resulting in marked changes in the cellular topology (plasma membrane blebbing).8,106

Most of the above-mentioned roles of actin in cellular biology apply to hepatocytes, and many of them are involved in biliary secretory processes. It is therefore not surprising that disorganization of the actin cytoskeleton induced by oxidative stress has several deleterious effects on hepatobiliary function.

The interrelationship between oxidative stress, Ca2+ elevations, actin-cytoskeletal integrity and hepatocanalicular secretory function was exhaustively investigated in the 90‘s by Prof. Coleman’s laboratory, using the hepatocyte couplet model. These studies revealed that, under mild oxidative stress conditions with preserved hepatocellular viability, a close relationship exists between the impairment in the capability of couplets to accumulate apically and retain in their canalicular vacuoles fluorescent bile-salt analogues and the disarrangement of the pericanalicular actin cytoskeleton induced by the oxidizing compounds, tBOOH98,107-109 and menadione.99,110,111 These two independent tests indicated that both the apical secretion of bile salts and their further tight-junctional-dependent retention in the bile canaliculus are impaired early under oxidative stress conditions. Essentially, the same findings were obtained in isolated perfused rat livers. When acutely administered in the perfusate at non-necrotic concentrations, tBOOH impaired bile salt excretion and increased the bile/perfusate ratio of [14C]- sucrose, a tight-junctional-integrity marker that enters the bile canaliculus via the paracellular pathway.70 The impairment in bile salt excretion and the disruption of the tight-junctional barrier function may be causally associated, since the latter induces leakage of bile salts (or other osmotically-active biliary solutes) from the canalicular space via the paracellular pathway, leading to dissipation of the osmotic gradients involved in bile formation.112

These functional alterations to secrete and retain bile salts in the biliary space seem to have a structural correlate. tBOOH induces disorganization of the tight-junctional complex in hepatocyte couplets, as suggested by the redistribution of the tight-junctional-associated protein, ZO-1.98 Changes in ZO-1 localization are however not observed in isolated perfused rat livers exposed to tBOOH, suggesting that the couplet model is a more sensitive model to study associations between functional and structural changes of the tight-junctional barrier. In addition, the canalicular bile salt transporter, Bsep, suffers endocytic internalization into intracellular vesicles in hepatocyte couplets,109 which reduces dramatically the density of transporters properly located at the membrane domain. A similar phenomenon has been described for Mrp2, another canalicular transporter relevant to bile formation. Indeed, retrieval of Mrp2 from the canalicular membrane into subapical vesicles occurs after exposure of isolated perfused rat livers to the pro-oxidant agents, tBOOH,71 chloro-dinitrobenzene71 and ethacrynic acid,75,113 or after hepatic ischemia-reperfusion.114 Mrp2 relocalization has a clear-cut functional correlate. Experiments in isolated perfused rat livers indicated that a high, sustained exposure to ethacrynic acid has an inhibitory effect on the excretion of both unchanged and conjugated forms of the model cholephilic dye, BSP.115 Haemodynamic changes induced by oxidative stress may also affect BSP hepatic handling, but the source of ROS should be extracellular rather than intracellular to induce this effect. Perfusion with a medium containing hypoxanthine in the presence of xanthine oxidase, which catalyzes in situ the conversion of hypoxanthine to O2•— and uric acid, increased hepatic vascular resistance in isolated rat livers, which is accompanied with a decrease in BSP uptake.116

The exact mechanisms that link oxidative-stress-induced actin disorganization with tight-junctional impairment and transporter internalization are unknown, but previous studies in the literature provide some clues. Hepatic tight-junctional permeability increases following actin-cytoskeleton disruption induced by phalloidin, a toxin that induces actin disorganization by binding specifically to F-actin and impeding its depolymerization.117 As indicated above, F-actin is anchored to tightjunctional-associated proteins (e.g., ZO-1, cingulin), and it is likely that F-actin-conformational disorganization induces relocalization of these intermediary proteins as well. In addition, F-actin disorganization may alter localization and/or occlusive function of proteins forming the tight-junctional strands, such as occludin and claudins. For example, ZO-1118 and cingulin119 bind to occludin apart from F-actin, and both tight-junctional-associated proteins interact with claudins as well.120 At present, the functional relevance of all these interactions remains speculative, and a clearer picture of the mechanisms involved in the actin-mediated oxidative tight-junctional impairment awaits further characterization.

In addition to impairing tight-junctional permeability, phalloidin-induced F-actin disorganization also induces internalization of canalicular transporters. This was shown to occur for Mrp2 and for one or more canalicular transporters belonging to the P-glycoprotein family (i.e., Mdr1a, Mdr1b, Mdr2 and Bsep).121 The retrieval of canalicular transporters under oxidative-stress conditions71,75,109,113 is therefore also likely due to the simultaneous F-actin disarrangement. The molecular bases to understand this causal relationship are just emerging. Mice lacking radixin, which links actin filaments to plasma-membrane proteins, develop conjugated hyperbilirubinemia associated to retrieval of Mrp2,122 and the same hols true for obstructive and estrogen-induced cholestasis, where a disturbed colocalization of Mrp2 and radixin is associated with Mrp2 endocytic internalization.123 Interestingly, the internalization of Mrp2 that occurs after hepatic ischemia-reperfusion is coincident with a virtual loss of radixin expression.124 Mrp2 retention in the apical membrane also requires interaction with the PDZ-domain protein, PDZK1.125 Since other canalicular transporters, such as Bsep, Mdr1 and Mdr2, do not contain obvious PDZ-interacting motifs, regulation by actin of the localization of these transporters must occur through a different subset of proteins. A likely candidate is HAX-1, a cytoskeleton-associated protein that interacts with the F-actin-binding protein, cortactin. HAX-1 has been recently identified as a binding partner for canalicular Bsep, Mdr1, and Mdr2.104 There is evidence that HAX-1 and cortactin participate in clathrin-mediated Bsep endocytosis from the canalicular plasma membrane,126 and that cortactin location in the cell cortex depends on an intact actin cytoskeleton.104 Again, further elucidation of the process regulating the interplay between canalicular transporters and actin-associated proteins should be directed to understand the mechanisms underlying the canalicular-transporter internalization under redox-imbalance conditions.

Finally, mechanisms other than actin disorganization should also be considered, and may well act in concert. Examples are the changes in cell volume due to oxidative stress. Loss of hepatocellular volume and retrieval of Mrp2 from the canalicular membrane occur concomitantly after addition of tBOOH71,127 or chloro-2,4-dinitrobenzene71 to isolated perfused rat livers, and hepatocyte shrinkage induced by hyperosmotic exposure in this experimental model triggers a similar retrieval.128 However, under these hyperosmotic conditions, distribution of the tight-junctional-associated protein, ZO-1, remains unaffected.128

3.3. Signal transduction pathways and oxidative stress-induced hepatocanalicular dysfunction: «The Ca2+-PKC Connection»

Elevation of cytosolic Ca2+ levels occurs under oxidative stress conditions. This is thought to involve both the entry of extracellular Ca2+ via plasma membrane receptor operated Ca2+ channels and the release of Ca2+ from intracellular Ca2+ storages, particularly in the endoplasmicreticulum (calciosome).129 Cytosolic Ca2+ elevation is a major determinant of the oxidative injury.130 As stated above, Ca2+ facilitates mitochondrial permeability transition, a key event leading to both necrosis and apoptosis. Besides, Ca2+ activates Ca2+-dependent proteases, endonucleases, and phospholipases, which reinforces mitochondrial mechanisms of cell death.11,15 However, more subtle changes in hepatocanalicular function can occur at lower, non-necrotic levels of oxidative stress, and Ca2+ elevations also seem to play a key role. The intracellular Ca2+ chelator, BAPTA/AM, fully prevented the impairment induced by low levels of tBOOH in the capability of the hepatocyte couplets to accumulate apically and retain in their canalicular vacuoles the bile salt-fluorescent analogue, CLF.110,111 Suggestively, the actin-cytoskeletal disarrangement is also prevented by this Ca2+-sequestering agent, further supporting a causal relationship between both phenomena. Furthermore, Ca2+-elevating agents, such as the Ca2+ ionophore A23187110 or the inhibitor of endoplasmic reticulum Ca2+-ATPase thapsigargin,70 mimic the deleterious effects of oxidative stress on both actin-cytoskeleton integrity and hepatocanalicular function. Taken together, these findings suggest that ROS-evoked Ca2+ signalling rather than a direct oxidation of actin plays a role in the secretory failure under mild oxidative-stress conditions.

Which are the signal pathways downstream of Ca2+ involved? Among the many ones regulating cell function, activation of Ca2+-dependent, «classical» protein kinase C isoforms (cPKCs) seems to be the more important. cPKCs contain one Ca2+-binding domain and, when cytosolic Ca2+ levels increase, recruitment of cPKCs to the plasma membrane takes place; this induces conformational changes of the enzyme leading to its activation.131 cPKC activation is a common feature under oxidative stress conditions.132 In particular, our group demonstrated that the pro-oxidant agent, tBOOH, induces cytosolic Ca2+ elevations and translocation of the cPKC isoform, PKCα, from the cytosol to the plasma membrane in isolated hepatocytes, even at concentrations low enough to only affect the biliary-secretory machinery.98 Furthermore, several previous findings showed strong similarities between the effect of oxidative stress and those induced by Ca2+ and PKC agonists, namely: i) Cytosolic Ca2+ elevations133 and PKC activation134 impaired bileflow generation in the isolated perfused rat liver,134 in part by increasing paracellular permeability;135-137 we and others further characterized these effects in the hepatocyte couplet model by showing that cytosolic Ca2+ elevations impair the couplet capability to secrete and retain in their canalicular vacuoles fluorescent bile salt analogues by activating cPKC,138 and that PKC activation by vasopressin and phorbol esters reproduced these effects.108,139,140ii) PKC agonists induce F-actin cytoskeletal disarrangements,139 and Ca2+-elevating agents reproduce these effects by a PKC-dependent mechanism.138

Final confirmation of a crucial role for cPKC activation in actin disorganization and the further impairment of hepatocanalicular function induced by ROS was provided by recent studies in hepatocyte couplets. ROS-mediated actin-cytoskeleton disarrangements were fully prevented by both PKC-pan-specific and cPKC-specific inhibitors.98 What is more relevant from the therapeutic point of view, both cytoskeleton disruption and canalicular dysfunction are reversed within 1 h by these inhibitors.98

The retrieval of the bile salt transporter, Bsep, from the canalicular membrane was also fully prevented by PKC antagonists.109 The same holds true for the impairment of the tight-junctional-retentive properties, another possible consequence of the actin disassembly induced by exposure to pro-oxidant agents.98 The exact mechanisms that explain the harmful effect of PKC on actin integrity and, by extension, to the hepatocanalicular function as a whole are unclear. Actin is a major target for PKC. This kinase phosphorylates and/or induces disorganization of a wide range of actin-cytoskeletal components, including actin itself, actin-associated proteins (e.g., α-actinin, vinculin and filamin), and membrane-cytoskeletal cross-linking proteins, including Rac (a member of the Rho family of small GTP-binding proteins), and myristoylated alaninerich C-kinase substrate (MARCKS).141,142 On the contrary, a direct phosphorylation of tight-junctional components mediated by cPKC is unlikely, since only «atypical» (non-Ca2+/diacylglycerol-dependent) PKC isoenzymes have been reported to concentrate at the tight-junctional complex.143 Alternatively, oxidative-stress-induced cPKC-dependent phosphorylation of components others than actin or actin-associated proteins is likely to contribute to Bsep retrieval. Selective activation of Ca2+-dependent PKCs by thymeleatoxin induces cholestasis and retrieval of Bsep from the canalicular membrane in the isolated perfused rat liver.144 Interestingly, the canalicular ATP-binding cassette transporter, multidrug resistance protein 1 (Mdr1), the closest homologue of Bsep, is phosphorylated by PKC at 3 serine residues at the C-terminal, «linker» region, which binds to the actin-associated protein, HAX-1.145 Whether this phosphorylation occurs also for Bsep remains to be ascertained.

The kind of canalicular protein that is internalized under oxidative-stress conditions and the signalling molecule involved seem to depends on the pro-oxidant agent employed, and on the magnitude of the oxidative damage. Low concentrations of the oxidizing compound, ethacrynic acid, does not translocate cPKC but «novel» PKC isoforms (nPKC). Under these conditions, the compound internalizes selectively Mrp2 without affecting Bsep, by a mechanism probably involving Ca2+-depending activation of inducible nitric oxide (NO) synthase (iNOS), followed by NO-mediated cGMP increase, and further cGMP-activated nPKC.113 However, higher ethacrynic acid doses, which can activate cPKC isoforms as well, induce internalization of both Bsep and Mrp2.113

As summarized in Figure 2, a picture is emerging to understand the effect of acutely-induced oxidative stress on the hepatobiliary function. Under mild ROS challenge not affecting hepatocellular viability, Ca2+ elevations induce cPKC and/or nPKC activation. This brings on a number of alterations in both function and localization of structures relevant to bile formation, such as actin cytoskeleton, canalicular transporters and tight-junctional components. This impairs, in turn, the biliary secretion and the further retention of solutes that provide osmotic driving force for bile formation. Other factors such as GSSG-induced cis-inhibition of the bile salt transport, reduced biliary excretion of glutathione due to intracellular glutathione depletion and hepatocellular death may become contributing factors depending on the magnitude of the pro-oxidant condition.

Figure 2.

Effect of the model oxidizing compound, tert-butylhydroperoxide (tBOOH), on the canalicular transport of monovalent bile salts (BS), excreted via the bile salt export pump (Bsep), and that of the multidrug-resistance associated protein 2 (Mrp2) substrate, oxidized glutathione (GSSG). In normal cells, the pericanalicular arrangement of the F-actin cytoskeleton allows for the appropriate insertion of the canalicular transporters in their membrane domain, and the proper barrier function of the tight-junctional structures. The acute administration of the oxidizing compound, tBOOH, stimulates formation of reactive oxygen species (ROS). This induces mobilization of Ca2+ across both the plasma and the calciosome membranes, and the subsequent activation of Ca2+-dependent («classical») PKC isoforms (cPKC). cPKC activation induces blebbing and redistribution of F-actin from the pericanalicular region to the cell body. This rearrangement, in turn, may lead to Bsep internalization and tight-junctional disorganization, which impairs BS/GSSG canalicular excretion, and their further retention inside the canalicular space. Ca2+ elevation may also activate inducible nitric oxide (NO) synthase (iNOS), which leads to NO-mediated guanylate cyclase (GC) activation and further cyclic guanosine monophosphate (cGMP)-mediated activation of «novel» PKC isoforms (nPKC). nPKC activation internalizes selectively Mrp2.

(0.18MB).
4. The antioxidant, adaptive hepatic response: «The ROS-busters»

In addition to the above-mentioned alterations induced acutely following the prooxidant insult, hepatocytes develop an adaptive response against oxidative stress, when the oxidative insult is maintained with time (Figure 3). This response involves induction of antioxidant defences such as catalase146 and manganese superoxide dismutase,147 a widespread phenomenon shared with other tissues. In addition, hepatic adaptation integrates its distinctive metabolizing and secretory capacity to reinforce this adaptive defence against the oxidative insult. Glutathione synthesis is increased by induction of γ-glutamyl-cysteinyl synthetase,148 and so are the phase-II-detoxifying enzymes, GST149 and UDP-glucuronosyltransferase.147 GST induction enhances the co-ordinated inactivation, via GSH conjugation, of DNA hydroperoxides and lipid hydroperoxides formed as secondary metabolites during oxidative stress.150 GST also catalyzes GSH conjugation of highly-reactive, toxic αβ-unsaturated lipid aldehydes generated by β-scission of lipid hydroperoxides of ω-6 polyunsaturated fatty acids, 4-hydroxy trans-2-nonenal (HNE) being the most abundant.151 These lipid-peroxidation products combine spontaneously with cysteine, histidine and lysine residues of proteins, potentially modifying protein function and resulting in cellular toxicity, and have been suggested to mediate and amplify the cellular effects of their free-radical precursors.152 UDP-glucuronosyltransferase induction would improve the metabolisation, via glucuronidation, of pro-oxidant toxicants, such as benzo(a)pyrene,153 pentachlorophenol,154 acetaminophen,155 aliphatic alcohols156 and manadione.157 Phase-II products are then extruded from the cell via the hepatocellular efflux pumps, MRP1, MRP2, MRP3, MRP4 (ABCC4) and the breast cancer resistance protein (BCRP, ABCG2), all of which are also upregulated.158-160 Glutathione conjugates are substrates of MRP2 and MRP1.161,162 Since these transporters also transfer GSSG, MRP1/2-mediated GSSG extrusion helps to maintain low intracellular GSSG levels, when GSSG reduction back to GSH via GSSG reductase becomes rate-limiting. Unlike MRP1, the basolateral carriers, MRP3, MRP4 and BCRP, transport non-glutathione conjugates, including glucurono-and sulpho-conjugates, and amidated bile salts.161,162 MRP1, MRP3 and MRP4 are normally expressed at very low levels in the basolateral membrane of the hepatocytes.161,162 Upregulation of basolateral extrusion pumps during a sustained oxidant insult is expected to shift the transfer of substrates normally excreted into bile towards blood, so that to permit urinary excretion. As an untoward effect of this adaptive response, this phenomenon might decrease the biliary excretion of bile salts, thus resulting in cholestasis. The extent at which this pathomechanism contributes to the cholestasis observed under sustained oxidative stress conditions (see bellow) remains to be ascertained.

Figure 3.

Main hepatocellular adaptive changes induced by sustained oxidative stress in the expression of enzymes and transporters involved in i) glutathione (GSH) synthesis via GSH synthetase (GS), ii) GSH-conjugation of lipid-peroxides or their aldehydic derivatives (LP) via glutathione 5-transferase (GST), and iii) plasmatic and biliary exportation of conjugated LPs and oxidized glutathione (GSSG) via both basolateral multidrug resistance-associated protein 1 (Mrpl) and canalicular multidrug resistance-associated protein 2 (Mrp2). When cells are exposed to oxidative stress, the transcription factor Nrf2, which is bound to the negative regulator Keapl in cytosol, escapes from its repression, and translocates and accumulates in the nucleus. Once there, it binds to the antioxidant response element (ARE) and activates ARE-dependent gene expression. Proteins that are induced by Nrf2 activation include γ-glutamyl-cysteinyl synthetase (γ-GCS), GST and Mrp2. Mrpl is induced by oxidative stress by a mechanism that seems to be independent of Nrf2. Only changes in rodent-gene products are displayed, since these regulations were mostly demonstrated in this species. For further details, see Section 4.

(0.12MB).

All these adaptive mechanisms are transcriptional in nature, and involve the coordinated activation of a number of redox-sensitive transcription factors. Early evidence that oxidative stress induces gene regulation came from the laboratories of Crawford and Cerutti,163 who reported on the induction of mRNA of the mouse protooncogenes, c-fos and c-myc, in JB6 cells by O2•— and H2O2. Soon later, a number of transcription factors, including Nrf2, NF-kB and AP-1, have been identified to be regulated by ROS, and to mediate expression of several gene products. Depending on the level of ROS, different redox-sensitive transcription factors are activated, and coordinate distinct biological responses. Low oxidative stress levels induce Nrf2,164 whereas intermediate levels of ROS trigger an inflammatory response through the activation of NF-kB and AP-1. High levels of oxidative stress surpass the defence capability of the antioxidant response, and induce mitochondrial permeability transition pores and disruption of the electron transfer, thereby resulting in apoptosis or necrosis.11

Nrf2 is a key transcription factor of the hepatic adaptation to sustained oxidative stress. Its induction has been linked to different oxidant agents such as the cancer chemoprotective agent, 3H-1,2-dimethiole-3-thione,147 alcohol,165 teributylhydroquinone,160 acetaminophen159 and bile salts.166 Activation of Nrf2 leads to the transcription of genes that codify phase II enzymes and hepatic transporters. Nrf2 induces the transcription of γ- glutamyl-cysteine synthetase,147 GST,147,149 UDPglucuronosyltransferase147,149,167 and the transporters, MRP2,158 MRP3,159 MRP4159 and BCRP;160 these upregulations were confirmed, in most cases, at the protein level. On the other hand, the role of Nrf2 in the increased expression of MRP1 at the protein level is not clear. In HepG2, induction is independent on this nuclear factor,160 whereas in knock-out mouse, the dependency is tissue-specific. For example, Nrf2 is required in fibroblast,168 but not in liver.159 These metabolic enzymes and carriers are also involved in the hepatic handling of electrophilic species that alkylate nucleophilic sites on peptides, proteins and nucleic acids, forming covalent adducts that can significantly compromise cellular integrity and function. Hence, Nrf2 is consider to be a sensor of, and a protector, against the so called «electrophilic stress».169,170

The action of Nrf2 depends on its accumulation in the nucleus, where it interacts with the antioxidant response element (ARE).171 This is a cis-acting enhancer sequence that contains the 5’-TGAC-3’ tetranucleotide present in the genes of enzymes associated with glutathione biosynthesis, redox proteins with active sulfhydryl moieties, drug-metabolizing enzymes and transporters. Nrf2 is a bZIP protein, and needs to form dimers in order to bind to ARE. It is not clear yet whether Nrf2 can interact with ARE as homodimer or it is obligated to form heterodimeric complexes with other bZIP proteins, like small RAF proteins.

The activation of Nrf2 has been suggested to be mediated by mechanisms that lead to its stabilization, increasing levels of cellular Nrf2 and subsequent transcriptional activity.172,173 Under basal conditions, Nrf2-dependent transcription is repressed by the negative regulator, Keap1. When cells are exposed to oxidative stress, electrophiles or chemopreventive agents, Nrf2 escapes Keap1-mediated repression, and translocates and accumulates in the nucleus, thus activating ARE-dependent gene expression to maintain cellular redox homeostasis.174,175 Several mechanisms have been proposed to explain the repressive effect of Keap1 on Nrf2, namely: i) Keap1 is a thiol-rich sensor protein, which is an adaptor protein for Cullin 3-dependent ubiquitination and degradation of Nrf2;176 electrophile modification of the Keap1 central linker domain would switch ubiquitination from Nrf2 to Keap1, leading to Nrf2 activation. ii) Upregulation of Nrf2 gene expression at the transcriptional level in response to ARE inducers.177iii) activation of PKC by oxidative stress and electrophiles which, in turn, activates Nrf2 through the phosphorylation of a serine residue located in the N-terminal region.178,179

5. Effect of chronic oxidative stress on hepatobiliary dysfunction: «The fight continues»

The adaptive, spontaneous mechanisms that take place in hepatocytes to minimize the deleterious effects of ROS is, however, not always sufficient to prevent hepatocellular oxidative damage. Under conditions in which ROS production is maintained at high levels during long periods, alterations in the capability of the hepatocyte to produce bile and to secrete cholephilic compounds have been described in the literature. However, the information is scarce, and cannot be unequivocally ascribed to the direct effect of ROS. Some of these changes may be secondary consequences of the oxidativestress-dependent inflammatory response that develops with time under long-standing pro-oxidant conditions, the hepatic accumulation of toxic biliary solutes due to the secretory failure (e.g. bile salts, bilirubin), or unspecific effects of the agents employed to cause the pro-oxidant injury.

The Long-Evans Cinnamon rat, which mimics Wilson’s disease, is a prototypical model of chronic hepatic oxidative stress. These rats exhibit inability to mobilize copper from the liver due to a genetic defect in the Atp7b gene, which encodes for a copper-transporting P-type ATPase. This transporter localizes to the trans-Golgi network or late endosomes under basal conditions, but it relocalizes in a copper-dependent manner to a subapical, vesicular compartment, which facilitates both copper incorporation into ceruloplasmin and biliary copper excretion via lysosomes.180 Apart from copper, Long-Evans Cinnamon rats also have abnormal hepatic accumulation of iron.181 Chronic copper/iron accumulation in these rats leads to a 50% increase in lipid peroxidation, presumably due to copper/iron capability to induce oxidative stress by generating ROS via both mitochondrial dysfunction182 and Fenton-type, copper/iron-catalyzed Haber-Weiss reaction.183,184 These mutant rats exhibit a reduced basal bile salt-biliary excretion compared with normal, Long-Evans Agouti rats. This alteration is linked to a low protein expression of the canalicular bile salt transporter, Bsep.185 A posttranscriptional mechanism seems to be involved, since no changes in the levels of Bsep mRNA has been observed.186 On the other hand, mRNA levels of the bile saltuptake systems, Ntcp and Oatps (Oatp1a1 and Oatp1a4 isoforms), are decreased, although this has not been confirmed at the protein level.186 Apart from alterations in bile salt hepatic handling, Long-Evans Cinnamon rats have hyperbilirubinaemia,184,187 and impairment of the excretion of the Mrp2 substrate, BSP.188 If these alterations involve changes in the expression of Mrp2, the rate-limiting step of both bilirubin and BSP hepatic handling, this would be nontranscriptional, since Mrp2 mRNA levels are normal in these rats.186 Alternatively, the above-mentioned transcriptional downregulation of Oatps, which also transport nonbile salt organic anions such as BSP and bilirubin, may be a contributing factor. Overall, these transcriptional and post-transcriptional alterations in transporter expression may help to explain why these rats have histological features of cholestasis.187

Another experimental model of metal-induced chronic oxidative stress is that afforded by long-term aluminum (Al3+) exposure to rats. Despite Al3+ is a non-redoxcation, it bears pro-oxidant activity. Al3+ facilitates O2•— formation induced by a number of pro-oxidant maneuvers; formation of the Al3+ superoxide radical ion, AlO2•2+, has been proposed to account for this property.189 This metal also facilitates iron-driven biological oxidative reactions.190 Finally, Al3+ induces mitochondrial permeability transition, which leads to ROS formation from mitochondrial origin.191 When administered intravenously for 1-2 weeks, Al3+ reduces bile flow, and this impairment correlates directly with Al3+ hepatic content; this was associated with elevations of serum bile salts, suggesting impaired hepatic handling of bile salts.192 A more chronic exposure to Al3+ (3 months), which doubles the lipid-peroxidation levels, also reduced bile flow and the biliary output of bile salts.90 Compartmental analysis of the plasma decay of BSP revealed that both sinusoidal uptake and canalicular excretion are decreased.90 The latter phenomenon was associated with a decrease in Mrp2 protein expression. All these alterations were prevented by administration of the antioxidant, vitamin E, suggesting that oxidative stress was the main, if not the only mechanism.89

Hepatic ischemia-reperfusion injury is another prototypical oxidative-stress mediated hepatopathy associated with both cholestasis21,87 and downregulation of hepatocellular transporters193,194(Figure 4). During the ischemic phase, the hepatic cells become more susceptible to oxygen toxicity during the reperfusion phase. This is due to multiple, self-aggravating mechanisms, mainly: i) mitochondrial permeability transition occurring by cytosolic Ca2+ elevations during the ischemic phase, which induces uncoupling of electrons from the transport chain when electron flow is restored by oxygen re-entry;195-197ii) proteolytic conversion of the enzyme, xanthine dehydrogenase, to the O2•—-generating enzyme, xanthine oxidase, by proteases released during the ischemic phase upon mitochondrial damage;197,198iii) cytokine-199 and complement-200 mediated activation of Kupffer cells, with further release of chemotactic and activating cytokines to neutrophils;201-203 both Kupffer cells and neutrophils can generate ROS, which further diffuse into hepatocytes.204,205

Figure 4.

Alterations of the expression of transporters relevant to bile formation in hepatic injury by ischemia-reperfusion. An ischemic period of 60 minutes followed by 1 day of reperfusion decreases the mRNA levels of the basolateral transporters Ntcp and Oatp (all isoforms), as well as those of the canalicular export transporters Mrp2 and Bsep. These transporter alterations have been confirmed at the protein level only for Mrp2. It is unknown whether ROS exert their effects at the transcriptional levels by directly activating certain redox-sensitive transcriptional factors (TF) or, indirectly, by promoting the release of cytokines as part of the inflammatory response secondary to the oxidative tissue injury which may, in turn, activate a different set of TFs. Since down-regulation of Mrp2 protein is more severe than that of mRNA, an additional post-transcriptional mechanism has been suggested, which may involve oxidative stress-induced transporter internalization, followed by delivery of the endocytosed transporters to the lysosomal compartment for degradation. Only changes in rodent-gene products are displayed, since these regulations were mostly demonstrated in this species. For further details, see Section 5.

(0.21MB).

Hepatic oxidative stress as a consequence of ischemiareperfusion depends on the duration of the ischemic phase.206,207 A 30-minute ischemia followed by different reperfusion periods either-in the isolated perfused rat liver model208 or in vivo86,207,209,210 does not affect both the GSSG hepatic content and/or the hepatic lipid-peroxidation levels, measured by generation of the secondary lipid-peroxidation by-products, such as malondialdehyde and HNE. This should not been interpreted as complete absence of oxidative stress, however, since the sensitivity of the methods measuring secondary lipid-peroxidation products is limited as compared with those measuring primary lipid peroxides,207,211,212 and the enzyme, glutathione reductase, may fully account for the conversion of GSSG back to GSH at low ROS levels;213 alternatively, ROS production may be too low to account for effective GSH conversion to GSSG.214 On the contrary, longer ischemic periods (45-90 minutes) followed by reperfusion result in increased lipid peroxidation, even assessing secondary lipid-peroxidation products.87,210,215-220

The impact of this maneuver on both bile-flow generation and the expression of transporters also depends on the duration of the ischemia. A 30-minute ischemia decreases both bile flow and biliary bile salt output after 1 day of reperfusion, but no changes in mRNA and protein levels of the basolateral and canalicular main bile salt transporters, Ntcp and Bsep, respectively, have been recorded.86 Functional/localization alterations of these transporters, or impairment of the expression of other bile salt transporters, such as Oatps, may be involved in the bile salt-secretory failure. Similarly, Mrp2 mRNA and protein expressions are unaffected, in agreement with the absence of changes in the maximum secretory rate of the Mrp2 substrate, ceftriaxone;86 lack of studies of ceftriaxone excretion under non-saturating conditions, however, does not allow changes in intrinsic transport efficiency to be ruled out. Unlike a 30-minute ischemia, a 60-minute ischemic period followed by 1 day of reperfusion decreases the mRNA levels of the basolateral transporters, Ntcp and Oatp (all isoforms), as well as those of the canalicular export transporters, Mrp2 and Bsep.193 This has been confirmed at the protein level for Mrp2.194 At least part of these transcriptional alterations may be due to the ROS-dependent hepatic inflammatory response, which leads to release of cholestatic, proinflammatory cytokines with capability to impair transporter expression at a transcriptional level.221 However, down-regulation of Mrp2 protein is more profound than that of mRNA, suggesting additional post-transcriptional mechanisms.194 Although the causes underlying the latter phenomenon are presently unknown, the early oxidative stress-induced transporter internalization, sustained with time, may lead to the delivery of the endocytosed transporters to the lysosomal compartment, followed by degradation, as was suggested to occur late in lipopolysaccharide-induced cholestasis222 and in obstructive cholestasis223 in rats. Apart from alterations in transporter expression/function, the tight-junctional barrier is impaired in ischemia-reperfusion injury, as shown in 24-or 48-h-could-stored isolated perfused rat livers subjected to reperfusion.224

Taken together, these models of long-lasting oxidative stress show consistently that cholestasis and/or impairment of the constitutive expression of transporters relevant to bile formation is a common feature in prolonged oxidative stress, and that both transcriptional and posttranscriptional mechanisms are involved. Proinflammatory cytokines released by the inflammatory response to the oxidative liver damage may be key mediators of part of these alterations.

6. Concluding remarks and future directions: «The end… of the beginning»

Oxidative stress is a cholestatic condition. Three consecutive phases in the oxidative injury affecting biliary secretion can be distinguished. Initially, the oxidative insult impairs acutely the hepatobiliary function, even at oxidative stress levels far lower than those affecting hepatocellular viability. These alterations comprise early impairment in hepatocyte capability to secrete and retain in the bile canaliculus osmotically-active solutes that are main determinants of bile generation, such as bile salts and GSH/GSSG. A key role for actin-cytoskeletal disarrangement on these functional alterations has been proposed, since it has been causally linked to internalization of canalicular transporters and disarrangement of tight-junctional structures. ROS-evoked, Ca2+-dependent signalling events, such as cPKC and/or nPKC activation, seem to mediate these harmful effects.

To limit these primary, deleterious consequences, hepatocytes develop a secondary adaptive response, triggered by ROS-responsive transcription factors, mainly the Nfr2/Keap1 system. This protective response involves up-regulations of the GSH-synthesizing enzyme, GSH-synthetase, the GSH-conjugating enzyme, GST, and the hepatocellular efflux pumps, Mrp2 and Mrp1. This enhances the co-ordinated inactivation of DNA/lipid hydroperoxides and their derivatives via GST-mediated GSH conjugation, and their further cellular extrusion via Mrp2 and Mrp1. Since, this adaptive response also involves upregulation of other basolateral pumps that are engaged in the transport of bile salts (Mrp3 and Mrp4), this may represent an additional mechanism of cholestasis, by shifting the transfer of these osmotically-active compounds from bile to blood.

Finally, a prolonged/high oxidative-stress challenge surpassing this adaptive antioxidant defence, such as that induced by ischemia-reperfusion or by accumulation of metals (e.g., Cu2+, Al3+), induces a number of alterations in hepatocellular transporters both at functional and expression levels. For the latter case, both transcriptional and posttranscriptional mechanisms have been reported, and the inflammatory response secondary to the oxidative tissue injury may play a key role.

Despite considerable progresses have been made in the characterization of the effects of oxidative stress on the biliary secretory machinery, the characterization of the molecular mechanisms underlying these effects is in its infancy. A bridge needs to be built between early events and late consequences of oxidative stress on bile secretion, in order to reconstruct the cascade of events leading to the posttranscriptional changes in transporter protein expression observed eventually during the sustained oxidative challenge. Also, we need to distinguish direct ROS-mediated effects from secondary consequences of the oxidative injury (e.g., inflammatory response, accumulation of biliary solutes). In addition, we must fully characterize the redox-sensitive signalling pathways involved in these effects; a more complete picture should provide more selective therapeutic strategies to interfere with ROS-mediated harmful pathways, or to enhance the protective ones. Studies on the impact of oxidative stress in transport function of biliary epithelial cells are also eagerly awaited; cholangiocytes may contribute to bile secretory failure, as they are the main target of cholangiopathies associated with periductal inflammation and oxidative stress. And last, but not least, it remains to be ascertained the actual contribution of oxidative stress in both transcriptional and posttranscriptional changes in liver transporter expression occurring in chronic cholestatic liver diseases in humans.161

Genomic and proteomic tools are accelerating the discovery of new ROS-responsive genes and the molecular targets of ROS action. These approaches are expected to greatly help to achieve the goals above. Meanwhile, we hope this preliminary information contributes to draw attention about the convenience of limiting oxidative stress in hepatopathies. Some short-scale clinical studies using co-adjuvant, antioxidant therapies have shown encouraging results,22 but multicentric and long-term clinical trials are needed to determine whether this strategy holds promise for the future.

Acknowledgements.

This work was supported by CONICET (PIP 6442) and Agencia Nacional de Promoción Científica y Tecnológica (ANPCyT; PICT 0526115), and Fundación Florencio Fiorini, Argentina.

References
[1.]
Nohl H, Gille L, Kozlov A, Staniek K..
Are mitochondria a spontaneous and permanent source of reactive oxygen species?.
Redox Rep, 8 (2003), pp. 135-141
[2.]
Cross A.R., Jones OT..
Enzymic mechanisms of superoxide production..
Biochim Biophys Acta, 1057 (1991), pp. 281-298
[3.]
Dey A, Cederbaum AI..
Alcohol and oxidative liver injury.
Hepatology, 43 (2006), pp. S63-74
[4.]
Kim K.B., Lee BM..
Oxidative stress to DNA, protein, and antioxidant enzymes (superoxide dismutase and catalase) in rats treated with benzo(a)pyrene.
Cancer Lett, 113 (1997), pp. 205-212
[5.]
Halliwell B..
Antioxidant defence mechanisms: from the beginning to the end (of the beginning).
Free Radic Res, 31 (1999), pp. 261-272
[6.]
Berlett B.S., Stadtman ER..
Protein oxidation in aging, disease, and oxidative stress.
J Biol Chem, 272 (1997), pp. 20313-20316
[7.]
Sohal RS..
Role of oxidative stress and protein oxidation in the aging process.
Free Radic Biol Med, 33 (2002), pp. 37-44
[8.]
Dalle-Donne I, Rossi R, Milzani A, Di Simplicio P, Colombo R..
The actin cytoskeleton response to oxidants: from small heat shock protein phosphorylation to changes in the redox state of actin itself.
Free Radic Biol Med, 31 (2001), pp. 1624-1632
[9.]
Bellomo G, Mirabelli F..
Oxidative stress and cytoskeletal alterations.
Ann N Y Acad Sci, 663 (1992), pp. 97-109
[10.]
Gores G.J., Herman B, Lemasters JJ..
Plasma membrane bleb formation and rupture: a common feature of hepatocellular injury.
Hepatology, 11 (1990), pp. 690-698
[11.]
Vercesi A.E., Kowaltowski A.J., Oliveira H.C., Castilho RF..
Mitochondrial Ca2+ transport, permeability transition and oxidative stress in cell death: implications in cardiotoxicity, neurodegeneration and dyslipidemias.
Front Biosci, 11 (2006), pp. 2554-2564
[12.]
Sawada M, Sester U, Carlson JC..
Superoxide radical formation and associated biochemical alterations in the plasma membrane of brain, heart, and liver during the lifetime of the rat.
J Cell Biochem, 48 (1992), pp. 296-304
[13.]
Konat GW..
H2O2-induced higher order chromatin degradation: a novel mechanism of oxidative genotoxicity.
J Biosci, 28 (2003), pp. 57-60
[14.]
Kovacic P, Jacintho JD..
Mechanisms of carcinogenesis: focus on oxidative stress and electron transfer.
Curr Med Chem, 8 (2001), pp. 773-796
[15.]
Ermak G, Davies KJ..
Calcium and oxidative stress: from cell signaling to cell death.
Mol Immunol, 38 (2002), pp. 713-721
[16.]
Cesaratto L, Vascotto C, Calligaris S, Tell G..
The importance of redox state in liver damage.
Ann Hepatol, 3 (2004), pp. 86-92
[17.]
Poli G..
Liver damage due to free radicals.
[18.]
Kaplowitz N, Tsukamoto H..
Oxidative stress and liver disease.
Prog Liver Dis, 14 (1996), pp. 131-159
[19.]
Medina J, Moreno-Otero R..
Pathophysiological basis for antioxidant therapy in chronic liver disease.
[20.]
Galaris D, Barbouti A, Korantzopoulos P..
Oxidative stress in hepatic ischemia-reperfusion injury: the role of antioxidants and iron chelating compounds.
Cur Pharm Des, 12 (2006), pp. 2875-2890
[21.]
Lemasters J.J., Thurman RG..
Reperfusion injury after liver preservation for transplantation.
Annu Rev Pharmacol Toxicol, 37 (1997), pp. 327-338
[22.]
Vendemiale G, Grattagliano I, Lupo L, Memeo V, Altomare E..
Hepatic oxidative alterations in patients with extra-hepatic cholestasis. Effect of surgical drainage.
J Hepatol, 37 (2002), pp. 601-605
[23.]
Sokol R.J., Devereaux M, Dahl R, Gumpricht E..
«Let there be bile»–understanding hepatic injury in cholestasis.
J Pediatr Gastroenterol Nutr, 43 (2006), pp. S4-9
[24.]
Harada K, Nakanuma Y..
Molecular mechanisms of cholangiopathy in primary biliary cirrhosis.
Med Mol Morphol, 39 (2006), pp. 55-61
[25.]
Salunga T.L., Cui Z.G., Shimoda S, Zheng H.C., Nomoto K, Kondo T, et al.
Oxidative stress-induced apoptosis of bile duct cells in primary biliary cirrhosis.
J Autoimmun, 29 (2007), pp. 78-86
[26.]
Aboutwerat A, Pemberton P.W., Smith A, Burrows P.C., McMahon R.F., Jain S.K., et al.
Oxidant stress is a significant feature of primary biliary cirrhosis.
Biochim Biophys Acta, 1637 (2003), pp. 142-150
[27.]
Salem T.A., El-Refaei M.F., Badra GA..
Study of antioxidant enzymes level and phagocytic activity in chronic liver disease patients.
Egypt J Immunol, 10 (2003), pp. 37-45
[28.]
Sakaguchi S, Furusawa S..
Oxidative stress and septic shock: metabolic aspects of oxygen-derived free radicals generated in the liver during endotoxemia.
FEMS Immunol Med Microbiol, 47 (2006), pp. 167-177
[29.]
Choi J, Ou JH..
Mechanisms of liver injury. III. Oxidative stress in the pathogenesis of hepatitis C virus.
Am J Physiol, 290 (2006), pp. G847-51
[30.]
Loguercio C, Federico A..
Oxidative stress in viral and alcoholic hepatitis.
Free Radic Biol Med, 34 (2003), pp. 1-10
[31.]
Stehbens WE..
Oxidative stress, toxic hepatitis, and antioxidants with particular emphasis on zinc.
Exp Mol Pathol, 75 (2003), pp. 265-276
[32.]
Pemberton P.W., Aboutwerat A, Smith A, Burrows P.C., McMahon R.F., Warnes TW..
Oxidant stress in type I autoimmune hepatitis: the link between necroinflammation and fibrogenesis?.
Biochim Biophys Acta, 1689 (2004), pp. 182-189
[33.]
Fernandez-Checa JC..
Alcohol-induced liver disease: when fat and oxidative stress meet.
Ann Hepatol, 2 (2003), pp. 69-75
[34.]
Gawrieh S, Opara E.C., Koch TR..
Oxidative stress in nonalcoholic fatty liver disease: pathogenesis and antioxidant therapies.
J Investig Med, 52 (2004), pp. 506-514
[35.]
Patrick L..
Nonalcoholic fatty liver disease: relationship to insulin sensitivity and oxidative stress. Treatment approaches using vitamin E, magnesium, and betaine.
Altern Med Rev, 7 (2002), pp. 276-291
[36.]
Albano E, Mottaran E, Occhino G, Reale E, Vidali M..
Review article: role of oxidative stress in the progression of non-alcoholic steatosis.
Aliment Pharmacol Ther, 22 (2005), pp. 71-73
[37.]
Papanikolaou G, Pantopoulos K..
Iron metabolism and toxicity.
Toxicol Appl Pharmacol, 202 (2005), pp. 199-211
[38.]
Alla V, Bonkovsky HL..
Iron in nonhemochromatotic liver disorders.
Semin Liver Dis, 25 (2005), pp. 461-472
[39.]
Ferenci P..
Pathophysiology and clinical features of Wilson disease.
Metab Brain Dis, 19 (2004), pp. 229-239
[40.]
Nanji A.A., Hiller-Sturmhofel S..
Apoptosis and necrosis: two types of cell death in alcoholic liver disease.
Alcohol Health Res World, 21 (1997), pp. 325-330
[41.]
Rosser B.G., Gores GJ..
Liver cell necrosis: cellular mechanisms and clinical implications.
Gastroenterology, 108 (1995), pp. 252-275
[42.]
Czaja MJ..
Induction and regulation of hepatocyte apoptosis by oxidative stress.
Antioxid Redox Signal, 4 (2002), pp. 759-767
[43.]
Higuchi H, Kurose I, Kato S, Miura S, Ishii H..
Ethanol induced apoptosis and oxidative stress in hepatocytes.
Alcohol Clin Exp Res, 20 (1996), pp. 340A-346A
[44.]
Cardin R, D’Errico A, Fiorentino M, Cecchetto A, Naccarato R, Farinati F..
Hepatocyte proliferation and apoptosis in relation to oxidative damage in alcohol-related liver disease.
Alcohol Alcohol, 37 (2002), pp. 43-48
[45.]
Anderson S, Nevins C.L., Green L.K., El-Zimaity H, Anand BS..
Assessment of liver histology in chronic alcoholics with and without hepatitis C virus infection.
Dig Dis Sci, 46 (2001), pp. 1393-1398
[46.]
Marinelli R.A., Gradilone S.A., Carreras F.I., Calamita G, Lehmann GL..
Liver aquaporins: significance in canalicular and ductal bile formation.
Ann Hepatol, 3 (2004), pp. 130-136
[47.]
Boyer JL..
New concepts of mechanisms of hepatocyte bile formation.
Physiol Rev, 60 (1980), pp. 303-326
[48.]
Elsing C, Kassner A, Hubner C, Buhli H, Stremmel W..
Absorptive and secretory mechanisms in biliary epithelial cells.
J Hepatol, 24 (1996), pp. 121-127
[49.]
Marinelli R.A., LaRusso NF..
Solute and water transport pathways in cholangiocytes.
Semin Liver Dis, 16 (1996), pp. 221-229
[50.]
Hagenbuch B, Stieger B, Foguet M, Lubbert H, Meier PJ..
Functional expression cloning and characterization of the hepatocyte Na+/bile acid cotransport system.
Proc Natl Acad Sci USA, 88 (1991), pp. 10629-10633
[51.]
Hagenbuch B, Meier PJ..
Molecular cloning, chromosomal localization, and functional characterization of a human liver Na+/bile acid cotransporter.
J Clin Invest, 93 (1994), pp. 1326-1331
[52.]
Bohan A, Boyer JL..
Mechanisms of hepatic transport of drugs: implications for cholestatic drug reactions.
Semin Liver Dis, 22 (2002), pp. 123-136
[53.]
Kouzuki H, Suzuki H, Ito K, Ohashi R, Sugiyama Y..
Contribution of sodium taurocholate co-transporting polypeptide to the uptake of its possible substrates into rat hepatocytes.
J Pharmacol Exp Ther, 286 (1998), pp. 1043-1050
[54.]
Kullak-Ublick G.A., Stieger B, Hagenbuch B, Meier PJ..
Hepatic transport of bile salts.
Semin Liver Dis, 20 (2000), pp. 273-292
[55.]
Hagenbuch B, Meier PJ..
The superfamily of organic anion transporting polypeptides.
Biochim Biophys Acta, 1609 (2003), pp. 1-18
[56.]
Pauli-Magnus C, Stieger B, Meier Y, Kullak-Ublick G.A., Meier PJ..
Enterohepatic transport of bile salts and genetics of cholestasis.
J Hepatol, 43 (2005), pp. 342-357
[57.]
Cui Y, Konig J, Leier I, Buchholz U, Keppler D..
Hepatic uptake of bilirubin and its conjugates by the human organic anion transporter SLC21A6.
J Biol Chem, 276 (2001), pp. 9626-9630
[58.]
Kamisako T, Kobayashi Y, Takeuchi K, Ishihara T, Higuchi K, Tanaka Y, et al.
Recent advances in bilirubin metabolism research: the molecular mechanism of hepatocyte bilirubin transport and its clinical relevance.
J Gastroenterol, 35 (2000), pp. 659-664
[59.]
Suchy F.J., Ananthanarayanan M..
Bile salt excretory pump: biology and pathobiology.
J Pediatr Gastroenterol Nutr, 43 (2006), pp. S10-6
[60.]
Nies A.T., Keppler D..
The apical conjugate efflux pump ABCC2 (MRP2).
Pflugers Arch, 453 (2007), pp. 643-659
[61.]
Ballatori N, Truong AT..
Relation between biliary glutathione excretion and bile acid-independent bile flow.
Am J Physiol, 256 (1989), pp. G22-30
[62.]
Ballatori N, Truong AT..
Glutathione as a primary osmotic driving force in hepatic bile formation.
Am J Physiol, 263 (1992), pp. G617-24
[63.]
Garcia-Ruiz C, Fernandez-Checa J.C., Kaplowitz N..
Bidirectional mechanism of plasma membrane transport of reduced glutathione in intact rat hepatocytes and membrane vesicles.
J Biol Chem, 267 (1992), pp. 22256-22264
[64.]
Ballatori N, Dutczak WJ..
Identification and characterization of high and low affinity transport systems for reduced glutathione in liver cell canalicular membranes.
J Biol Chem, 269 (1994), pp. 19731-19737
[65.]
Yang B, Hill CE..
Nifedipine modulation of biliary GSH and GSSG/conjugate efflux in normal and regenerating rat liver.
Am J Physiol, 281 (2001), pp. G85-94
[66.]
Inoue M, Kinne R, Tran T, Arias IM..
The mechanism of biliary secretion of reduced glutathione. Analysis of transport process in isolated rat liver canalicular membrane vesicles.
Eur J Biochem, 134 (1983), pp. 467-471
[67.]
Fernandez-Checa J.C., Takikawa H, Horie T, Ookhtens M, Kaplowitz N..
Canalicular transport of reduced glutathione in normal and mutant Eisai hyperbilirubinemic rats.
J Biol Chem, 267 (1992), pp. 1667-1673
[68.]
Bogert P.T., LaRusso NF..
Cholangiocyte biology.
Curr Opin Gastroenterol, 23 (2007), pp. 299-305
[69.]
Xia X, Francis H, Glaser S, Alpini G, Lesage G..
Bile acid interactions with cholangiocytes.
World J Gastroenterol, 12 (2006), pp. 3553-3563
[70.]
Ballatori N, Truong AT..
Altered hepatic junctional permeability, bile acid excretion and glutathione efflux during oxidant challenge.
J Pharmacol Exp Ther, 251 (1989), pp. 1069-1075
[71.]
Schmitt M, Kubitz R, Wettstein M, vom Dahl S, Haussinger D..
Retrieval of the mrp2 gene encoded conjugate export pump from the canalicular membrane contributes to cholestasis induced by tert-butyl hydroperoxide and chloro-dinitrobenzene.
Biol Chem, 381 (2000), pp. 487-495
[72.]
Akerboom T.P., Bilzer M, Sies H..
Relation between glutathione redox changes and biliary excretion of taurocholate in perfused rat liver.
J Biol Chem, 259 (1984), pp. 5838-5843
[73.]
te Koppele J.M., Keller B.J., Caldwell-Kenkel J.C., Lemasters J.J., Thurman RG..
Effect of hepatotoxic chemicals and hypoxia on hepatic nonparenchymal cells: impairment of phagocytosis by Kupffer cells and disruption of the endothelium in rat livers perfused with colloidal carbon.
Toxicol Appl Pharmacol, 110 (1991), pp. 20-30
[74.]
Eipel C, Eisold M, Schuett H, Vollmar B..
Inhibition of heme oxygenase-1 protects against tissue injury in carbon tetrachloride exposed livers.
J Surg Res, 139 (2007), pp. 113-120
[75.]
Ji B, Ito K, Sekine S, Tajima A, Horie T..
Ethacrynic-acid-induced glutathione depletion and oxidative stress in normal and Mrp2-deficient rat liver.
Free Radic Biol Med, 37 (2004), pp. 1718-1729
[76.]
Barros S.B., Videla L.A., Simizu K, Van H.L., Junqueira VB..
Lindane-induced oxidative stress. II. Time course of changes in hepatic glutathione status.
Xenobiotica, 18 (1988), pp. 1305-1310
[77.]
Bramow S, Ott P, Thomsen N.F., Bangert K, Tygstrup N, Dalhoff K..
Cholestasis and regulation of genes related to drug metabolism and biliary transport in rat liver following treatment with cyclosporine A and sirolimus (Rapamycin).
Pharmacol Toxicol, 89 (2001), pp. 133-139
[78.]
Veggi L.M., Crocenzi F.A., Roma M.G., Dawson P.A., Pellegrino J.M., Sanchez Pozzi E.J., et al.
Dapsone-induced cholestasis and impairment of bile salt output in the rat.
Biochem Pharmacol, 63 (2002), pp. 1553-1563
[79.]
Veggi L.M., Crocenzi F.A., Roma M.G., Mottino AD..
Dapsone impairs the bile salt-independent fraction of bile flow in rats: Possible involvement of its N-hydroxylated metabolite.
Toxicology, 211 (2005), pp. 97-106
[80.]
Hoener B, Noach A, Andrup M, Yen TS..
Nitrofurantoin produces oxidative stress and loss of glutathione and protein thiols in the isolated perfused rat liver.
Pharmacology, 38 (1989), pp. 363-373
[81.]
Hoener BA..
Nitrofurazone: kinetics and oxidative stress in the singlepass isolated perfused rat liver.
Biochem Pharmacol, 37 (1988), pp. 1629-1636
[82.]
Settaf A, Zahidy M, Elimadi A, Sapena R, Alsamad I.A., Tillement J, et al.
S-15176 reduces the hepatic injury in rats subjected to experimental ischemia and reperfusion.
Eur J Pharmacol, 406 (2000), pp. 281-292
[83.]
Koeppel T.A., Trauner M, Mennone A, Arrese M, Rios-Velez L, Boyer JL..
Role of glutathione in hepatic bile formation during reperfusion after cold ischemia of the rat liver.
J Hepatol, 28 (1998), pp. 812-819
[84.]
Karwinski W, Soreide O..
Allopurinol improves scavenging ability of the liver after ischemia/reperfusion injury.
Liver, 17 (1997), pp. 139-143
[85.]
Bowers B.A., Branum G.D., Rotolo F.S., Watters C.R., Meyers WC..
Bile flow-an index of ischemic injury..
J Surg Res, 42 (1987), pp. 565-569
[86.]
Accatino L, Pizarro M, Solis N, Arrese M, Koenig CS..
Bile secretory function after warm hepatic ischemia-reperfusion injury in the rat.
Liver Transpl, 9 (2003), pp. 1199-1210
[87.]
Lee S.M., Park M.J., Cho T.S., Clemens MG..
Hepatic injury and lipid peroxidation during ischemia and reperfusion.
Shock, 13 (2000), pp. 279-284
[88.]
Turnage R.H., Bagnasco J, Berger J, Guice K.S., Oldham K.T., Hinshaw DB..
Hepatocellular oxidant stress following intestinal ischemia-reperfusion injury.
J Surg Res, 51 (1991), pp. 467-471
[89.]
Gonzalez M.A., Alvarez M.L., Pisani G.B., Bernal C.A., Roma M.G., Carrillo MC..
Involvement of oxidative stress in the impairment in biliary secretory function induced by intraperitoneal administration of aluminum to rats.
Biol Trace Elem Res, 116 (2007), pp. 329-348
[90.]
Gonzalez M.A., Roma M.G., Bernal C.A., Alvarez M.L., Carrillo MC..
Biliary secretory function in rats chronically intoxicated with aluminum.
Toxicol Sci, 79 (2004), pp. 189-195
[91.]
Lauterburg B.H., Smith C.V., Hughes H, Mitchell JR..
Biliary excretion of glutathione and glutathione disulfide in the rat Regulation and response to oxidative stress.
J Clin Invest, 73 (1984), pp. 124-133
[92.]
Akerboom T, Bultmann T, Sies H..
Inhibition of biliary taurocholate excretion during menadione metabolism in perfused rat liver.
Arch Biochem Biophys, 263 (1988), pp. 10-18
[93.]
Chenderovitch J, Raizman A, Infante R..
Mechanism of ethacrynic acid-induced choleresis in the rat.
Am J Physiol, 229 (1975), pp. 1180-1187
[94.]
Singh R, Czaja MJ..
Regulation of hepatocyte apoptosis by oxidative stress.
J Gastroenterol Hepatol, 22 (2007), pp. S45-8
[95.]
Kim J.S., He L, Lemasters JJ..
Mitochondrial permeability transition: a common pathway to necrosis and apoptosis.
Biochem Biophys Res Commun, 304 (2003), pp. 463-470
[96.]
Nieminen AL..
Apoptosis and necrosis in health and disease: role of mitochondria.
Int Rev Cytol, 224 (2003), pp. 29-55
[97.]
Griffiths J.C., Sies H, Meier P.J., Akerboom TP..
Inhibition of taurocholate efflux from rat hepatic canalicular membrane vesicles by glutathione disulfide.
FEBS Lett, 213 (1987), pp. 34-38
[98.]
Pérez L.M., Milkiewicz P, Ahmed-Choudhury J, Elias E, Ochoa J.E., Sanchez Pozzi E.J., et al.
Oxidative stress induces actincytoskeletal and tight-junctional alterations in hepatocytes by a Ca2+-dependent, PKC-mediated mechanism: protective effect of PKA.
Free Radic Biol Med, 40 (2006), pp. 2005-2017
[99.]
Stone V, Coleman R, Chipman JK..
Comparison of the effects of redox cycling and arylating quinones on hepatobiliary function and glutathione homeostasis in rat hepatocyte couplets.
Toxicol Appl Pharmacol, 138 (1996), pp. 195-200
[100.]
Small J.V., Rottner K, Kaverina I..
Functional design in the actin cytoskeleton.
Curr Opin Cell Biol, 11 (1999), pp. 54-60
[101.]
Anderson J.M., Van Itallie CM..
Tight junctions and the molecular basis for regulation of paracellular permeability.
Am J Physiol, 269 (1995), pp. G467-75
[102.]
Bi G.Q., Morris R.L., Liao G, Alderton J.M., Scholey J.M., Steinhardt RA..
Kinesin-and myosin-driven steps of vesicle recruitment for Ca2+-regulated exocytosis.
J Cell Biol, 138 (1997), pp. 999-1008
[103.]
Kato Y, Watanabe C, Tsuji A..
Regulation of drug transporters by PDZ adaptor proteins and nuclear receptors.
Eur J Pharm Sci, 27 (2006), pp. 487-500
[104.]
Ortiz D.F., Moseley J, Calderon G, Swift A.L., Li S, Arias IM..
Identification of HAX-1 as a protein that binds bile salt export protein and regulates its abundance in the apical membrane of Madin-Darby canine kidney cells.
J Biol Chem, 279 (2004), pp. 32761-32770
[105.]
Gallagher A.R., Cedzich A, Gretz N, Somlo S, Witzgall R..
The polycystic kidney disease protein PKD2 interacts with Hax-1, a protein associated with the actin cytoskeleton.
Proc Natl Acad Sci USA, 97 (2000), pp. 4017-4022
[106.]
Mirabelli F, Salis A, Marinoni V, Finardi G, Bellomo G, Thor H, et al.
Menadione-induced bleb formation in hepatocytes is associated with the oxidation of thiol groups in actin.
Arch Biochem Biophys, 264 (1988), pp. 261-269
[107.]
Ahmed-Choudhury J, Orsler D.J., Coleman R..
Hepatobiliary effects of tertiary-butylhydroperoxide tBOOH) in isolated rat hepatocyte couplets.
Toxicol Appl Pharmacol, 152 (1998), pp. 270-275
[108.]
Roma M.G., Orsler D.J., Coleman R..
Canalicular retention as a marker of tight junctional permeability in isolated hepatocyte couplets: effects of protein kinase modulation and cholestatic agents.
Fund Appl Toxicol, 37 (1997), pp. 71-81
[109.]
Pérez L.M., Milkiewicz P, Elias E, Coleman R, Sanchez Pozzi E.J., Roma MG..
Oxidative stress induces internalization of the bile salt export pump, Bsep, and bile salt secretory failure in isolated rat hepatocyte couplets: a role for protein kinase C and prevention by protein kinase A.
Toxicol Sci, 91 (2006), pp. 150-158
[110.]
Stone V, Johnson G.D., Wilton J.C., Coleman R, Chipman JK..
Effect of oxidative stress and disruption of Ca2+ homeostasis on hepatocyte canalicular function.
in vitro. Biochem Pharmacol, 47 (1994), pp. 625-632
[111.]
Stone V, Lankester D.J., Wilton J.C., Coleman R, Chipman JK..
Toxicological insult of analicular function in rat hepatocyte couplets: the role of calcium.
Toxicol in Vitro, 8 (1994), pp. 539-541
[112.]
Boyer JL..
Tight junctions in normal and cholestatic liver: does the paracellular pathway have functional significance?.
Hepatology, 3 (1983), pp. 614-617
[113.]
Sekine S, Ito K, Horie T..
Oxidative stress and Mrp2 internalization.
Free Radic Biol Med, 40 (2006), pp. 2166-2174
[114.]
Yu Q.Y., Shu M, Dai J.H., Ma J.B., Yu Y, Liu DH..
The mechanism of the increase of plasma bilirubin after hepatic ischemiareperfusion in rats.
Zhonghua Gan Zang Bing Za Zhi, 15 (2007), pp. 763-766
[115.]
James S.I., Ahokas JT..
Modulation of sulphobromophthalein excretion by ethacrynic acid.
Xenobiotica, 22 (1992), pp. 1433-1439
[116.]
Kawamoto S, Tashiro S, Miyauchi Y, Inoue M..
Changes in circulatory status and transport function of the liver induced by reactive oxygen species.
Am J Physiol, 268 (1995), pp. G47-53
[117.]
Elias E, Hruban Z, Wade J.B., Boyer JL..
Phalloidin-induced cholestasis: a microfilament-mediated change in junctional complex permeability.
Proc Natl Acad Sci USA, 77 (1980), pp. 2229-2233
[118.]
Fanning A.S., Jameson B.J., Jesaitis L.A., Anderson JM..
The tight junction protein ZO-1 establishes a link between the transmembrane protein occludin and the actin cytoskeleton.
J Biol Chem, 273 (1998), pp. 29745-29753
[119.]
D’Atri F, Citi S..
Cingulin interacts with F-actin in vitro.
FEBS Lett, 507 (2001), pp. 21-24
[120.]
Heiskala M, Peterson P.A., Yang Y..
The roles of claudin super-family proteins in paracellular transport.
Traffic, 2 (2001), pp. 93-98
[121.]
Rost D, Kartenbeck J, Keppler D..
Changes in the localization of the rat canalicular conjugate export pump Mrp2 in phalloidin-induced cholestasis.
Hepatology, 29 (1999), pp. 814-821
[122.]
Kocher O, Comella N, Gilchrist A, Pal R, Tognazzi K, Brown L.F., et al.
PDZK1, a novel PDZ domain-containing protein upregulated in carcinomas and mapped to chromosome 1q21, interacts with cMOAT (MRP2), the multidrug resistance-associated protein.
Lab Invest, 79 (1999), pp. 1161-1170
[123.]
Kojima H, Sakurai S, Yoshiji H, Uemura M, Yoshikawa M, Fukui H..
The role of radixin in altered localization of canalicular conjugate export pump Mrp2 in cholestatic rat liver.
Hepatol Res, 38 (2008), pp. 202-210
[124.]
Shu M, Peng C.H., Chen H, Shen B.Y., Qiu W.H., Jiang Z.H., et al.
Expression and localization of multi-drug resistance-associated protein 2 and radixin after hepatic ischemia-reperfusion: experiment with rats.
Zhonghua Yi Xue Za Zhi, 87 (2007), pp. 947-952
[125.]
Kikuchi S, Hata M, F.K. Yamane, Y Matsui, T Tamura A, et al.
Radixin deficiency causes conjugated hyperbilirubinemia with loss of Mrp2 from bile canalicular membranes.
Nat Genet, 31 (2002), pp. 320-325
[126.]
Wu H, Parsons JT..
Cortactin, an 80/85-kilodalton pp60src substrate, is a filamentous actin-binding protein enriched in the cell cortex.
J Cell Biol, 120 (1993), pp. 1417-1426
[127.]
Hallbrucker C, Ritter M, Lang F, Gerok W, Haussinger D..
Hydroperoxide metabolism in rat liver. K+ channel activation, cell volume changes and eicosanoid formation.
Eur J Biochem, 211 (1993), pp. 449-458
[128.]
Kubitz R, D’Urso D, Keppler D, Häussinger D..
Osmodependent dynamic localization of the multidrug resistance protein 2 in the rat hepatocyte canalicular membrane.
Gastroenterology, 113 (1997), pp. 1438-1442
[129.]
Reed DJ..
Status of calcium and thiols in hepatocellular injury by oxidative stress.
Semin Liver Dis, 10 (1990), pp. 285-292
[130.]
Orrenius S, Burkitt M.J., Kass G.E., Dypbukt J.M., Nicotera P..
Calcium ions and oxidative cell injury.
Ann Neurol, 32 (1992), pp. S33-42
[131.]
Shirai Y, Saito N..
Activation mechanisms of protein kinase C: maturation, catalytic activation, and targeting.
J Biochem (Tokyo), 132 (2002), pp. 663-668
[132.]
Gopalakrishna R, Jaken S..
Protein kinase C signaling and oxidative stress.
Free Radic Biol Med, 28 (2000), pp. 1349-1361
[133.]
Nathanson M.H., Gautam A, Bruck R, Isales C.M., Boyer JL..
Effects of Ca2+ agonists on cytosolic Ca2+ in isolated hepatocytes and on bile secretion in the isolated perfused rat liver.
Hepatology, 15 (1992), pp. 107-116
[134.]
Corasanti J.G., Smith N.D., Gordon E.R., Boyer JL..
Protein kinase C agonists inhibit bile secretion independently of effects on the microcirculation in the isolated perfused rat liver.
Hepatology, 10 (1989), pp. 8-13
[135.]
Kan K.S., Coleman R..
The calcium ionophore A23187 increases the tight-junctional permeability in rat liver.
Biochem J, 256 (1988), pp. 1039-1041
[136.]
Lowe P.J., Miyai K, Steinbach J.H., Hardison WG..
Hormonal regulation of hepatocyte tight junctional permeability.
Am J Physiol, 255 (1988), pp. G454-61
[137.]
Llopis J, Kass G.E., Duddy S.K., Farell G.C., Gahm A, Orrenius S..
Mobilization of the hormone-sensitive calcium pool increases hepatocyte tight junctional permeability in the perfused rat liver.
FEBS Lett, 280 (1991), pp. 84-86
[138.]
Roma M.G., Ahmed-Choudhury J, Coleman R..
The protein kinase inhibitor 1-(5-isoquinolinylsulfonyl)-2-methyl-piperzine (H-7) prevents and reverses Ca2+-mediated injury in isolated rat hepatocyte couplets.
Toxicol Appl Pharmacol, 161 (1999), pp. 192-201
[139.]
Roma M.G., Stone V, Shaw R, Coleman R..
Vasopressin-induced disruption of actin cytoskeletal organization and canalicular function in isolated rat hepatocyte couplets: possible involvement of protein kinase C.
Hepatology, 28 (1998), pp. 1031-1041
[140.]
Nathanson M.H., Gautam A, Ng O.C., Bruck R, Boyer JL..
Hormonal regulation of paracellular permeability in isolated rat hepatocyte couplets.
Am J Physiol, 262 (1992), pp. G1079-86
[141.]
Keenan C, Kelleher D..
Protein kinase C and the cytoskeleton.
Cell Signal, 10 (1998), pp. 225-232
[142.]
Larsson C..
Protein kinase C and the regulation of the actin cytoskeleton.
Cell Signal, 18 (2006), pp. 276-284
[143.]
Dodane V, Kachar B..
Identification of isoforms of G proteins and PKC that colocalize with tight junctions.
J Membr Biol, 149 (1996), pp. 199-209
[144.]
Kubitz R, Saha N, Kuhlkamp T, Dutta S, vom Dahl S, Wettstein M, et al.
Ca2+-dependent protein kinase C-isoforms induce cholestasis in rat liver.
J Biol Chem, 279 (2004), pp. 10323-10330
[145.]
Chambers T.C., Pohl J, Raynor R.L., Kuo JF..
Identification of specific sites in human P-glycoprotein phosphorylated by protein kinase C..
J Biol Chem, 268 (1993), pp. 4592-4595
[146.]
Sen P, Chakraborty P.K., Raha S..
p38 mitogen-activated protein kinase (p38MAPK) upregulates catalase levels in response to low dose H2O2 treatment through enhancement of mRNA stability.
FEBS Lett, 579 (2005), pp. 4402-4406
[147.]
Kwak M.K., Itoh K, Yamamoto M, Sutter T.R., Kensler TW..
Role of transcription factor Nrf2 in the induction of hepatic phase 2 and antioxidative enzymes in vivo by the cancer chemoprotective agent, 3H-1, 2-dimethiole-3-thione.
Mol Med, 7 (2001), pp. 135-145
[148.]
Yamane Y, Furuichi M, Song R, Van N.T., Mulcahy R.T., Ishikawa T, et al.
Expression of multidrug resistance protein/GS-X pump and gamma-glutamylcysteine synthetase genes is regulated by oxidative stress.
J Biol Chem, 273 (1998), pp. 31075-31085
[149.]
Kohle C, Bock KW..
Coordinate regulation of Phase I and II xenobiotic metabolisms by the Ah receptor and Nrf2.
Biochem Pharmacol, 73 (2007), pp. 1853-1862
[150.]
Ketterer B, Meyer DJ..
Glutathione transferases: a possible role in the detoxication and repair of DNA and lipid hydroperoxides.
Mutat Res, 214 (1989), pp. 33-40
[151.]
Renes J, de Vries E.E., Hooiveld G.J., Krikken I, Jansen P.L., Muller M..
Multidrug resistance protein MRP1 protects against the toxicity of the major lipid peroxidation product 4-hydroxynonenal.
Biochem J, 350 (2000), pp. 555-561
[152.]
Srivastava S, Chandra A, Wang L.F., Seifert W.E. Jr., DaGue B.B., Ansari N.H., et al.
Metabolism of the lipid peroxidation product, 4-hydroxy-trans-2-nonenal, in isolated perfused rat heart.
J Biol Chem, 273 (1998), pp. 10893-10900
[153.]
Byczkowski J.Z., Gessner T..
Effects of superoxide generated in vitro on glucuronidation of benzo[a]pyrene metabolites by mouse liver microsomes.
Int J Biochem, 19 (1987), pp. 531-537
[154.]
Umemura T, Kuroiwa Y, Kitamura Y, Ishii Y, Kanki K, Kodama Y, et al.
A crucial role of Nrf2 in in vivo defense against oxidative damage by an environmental pollutant, pentachlorophenol.
Toxicol Sci, 90 (2006), pp. 111-119
[155.]
Clement Y.N., Williams AF..
Protection against paracetamol-induced hepatic injury by prazosin pre-treatment in CD-1 mice.
Mutat Res, 579 (2005), pp. 182-188
[156.]
Ebner T, Burchell B..
Substrate specificities of two stably expressed human liver UDP-glucuronosyltransferases of the UGT1 gene family.
Drug Metab Dispos, 21 (1993), pp. 50-55
[157.]
Liu R.M., Sainsbury M, Tabor M.W., Shertzer HG..
Mechanisms of protection from menadione toxicity by 5,10-dihydroindeno[1,2,-b]indole in a sensitive and resistant mouse hepatocyte line.
Biochem Pharmacol, 46 (1993), pp. 1491-1499
[158.]
Vollrath V, Wielandt A.M., Iruretagoyena M, Chianale J..
Role of Nrf2 in the regulation of the Mrp2 (ABCC2) gene.
Biochem J, 395 (2006), pp. 599-609
[159.]
Aleksunes L.M., Slitt A.L., Maher J.M., Augustine L.M., Goedken M.J., Chan J.Y., et al.
Induction of Mrp3 and Mrp4 transporters during acetaminophen hepatotoxicity is dependent on Nrf2.
Toxicol Appl Pharmacol, 226 (2008), pp. 74-83
[160.]
Adachi T, Nakagawa H, Chung I, Hagiya Y, Hoshijima K, Noguchi N, et al.
Nrf2-dependent and-independent induction of ABC transporters ABCC1, ABCC2, and ABCG2 in HepG2 cells under oxidative stress.
J Exp Ther Oncol, 6 (2007), pp. 335-348
[161.]
Geier A, Wagner M, Dietrich C.G., Trauner M..
Principles of hepatic organic anion transporter regulation during cholestasis, inflammation and liver regeneration.
Biochim Biophys Acta, 1773 (2007), pp. 283-308
[162.]
Homolya L, Varadi A, Sarkadi B..
Multidrug resistance-associated proteins: export pumps for conjugates with glutathione, glucuronate or sulfate.
Biofactors, 17 (2003), pp. 103-114
[163.]
Crawford D.R., Cerutti PA..
Expression of oxidant stress-related genes in tumor promotion of mouse epidermal JB6 cells., New York Plenum Press, (1987), pp. 183-190
[164.]
Halliwell B, Gutteridge J..
Free radicals in biology and medicine., Oxford Oxford University Press, (1999),
[165.]
Gong P, Cederbaum AI..
Nrf2 is increased by CYP2E1 in rodent liver and HepG2 cells and protects against oxidative stress caused by CYP2E1.
Hepatology, 43 (2006), pp. 144-153
[166.]
Tan K.P., Yang M, Ito S..
Activation of nuclear factor (erythroid-2 like) factor 2 by toxic bile acids provokes adaptive defense responses to enhance cell survival at the emergence of oxidative stress.
Mol Pharmacol, 72 (2007), pp. 1380-1390
[167.]
Yueh M.F., Tukey RH..
Nrf2-Keap1 signaling pathway regulates human UGT1A1 expression. in vitro and in transgenic UGT1 mice.
J Biol Chem, 282 (2007), pp. 8749-8758
[168.]
Hayashi A, Suzuki H, Itoh K, Yamamoto M, Sugiyama Y..
Transcription factor Nrf2 is required for the constitutive and inducible expression of multidrug resistance-associated protein1 in mouse embryo fibroblasts.
Biochem Biophys Res Commun, 310 (2003), pp. 824-829
[169.]
Wakabayashi N, nkova-Kostova A.T., Holtzclaw W.D., Kang M.I., Kobayashi A, Yamamoto M, et al.
Protection against electrophile and oxidant stress by induction of the phase 2 response: fate of cysteines of the Keap1 sensor modified by inducers.
Proc Natl Acad Sci USA, 101 (2004), pp. 2040-2045
[170.]
Maher J.M., Dieter M.Z., Aleksunes L.M., Slitt A.L., Guo G, Tanaka Y, et al.
Oxidative and electrophilic stress induces multidrug resistance-associated protein transporters via the nuclear factor-E2-related factor-2 transcriptional pathway.
Hepatology, 46 (2007), pp. 1597-1610
[171.]
Nguyen T, Sherratt P.J., Pickett CB..
Regulatory mechanisms controlling gene expression mediated by the antioxidant response element.
Annu Rev Pharmacol Toxicol, 43 (2003), pp. 233-260
[172.]
Nguyen T, Sherratt P.J., Huang H.C., Yang C.S., Pickett CB..
Increased protein stability as a mechanism that enhances Nrf2-mediated transcriptional activation of the antioxidant response element: degradation of Nrf2 by the 26 S proteasome.
J Biol Chem, 278 (2003), pp. 4536-4541
[173.]
Stewart D, Killeen E, Naquin R, Alam S, Alam J..
Degradation of transcription factor Nrf2 via the ubiquitin-proteasome pathway and stabilization by cadmium.
J Biol Chem, 278 (2003), pp. 2396-2402
[174.]
Zhang DD..
Mechanistic studies of the Nrf2-Keap1 signaling pathway.
Drug Metab Rev, 38 (2006), pp. 769-789
[175.]
Itoh K, Wakabayashi N, Katoh Y, Ishii T, Igarashi K, Engel J.D., et al.
Keap1 represses nuclear activation of antioxidant responsive elements by Nrf2 through binding to the amino-terminal Neh2 domain.
Genes Dev, 13 (1999), pp. 76-86
[176.]
Hong F, Sekhar K.E., Freeman M.L., Liebler DC..
Specific patterns of electrophile adduction trigger Keap1 ubiquitination and Nrf2 activation.
J Biol Chem, 280 (2005), pp. 31768-31775
[177.]
Kwak M.K., Itoh K, Yamamoto M, Kensler TW..
Enhanced expression of the transcription factor Nrf2 by cancer chemopreventive agents: Role of antioxidant response element-like sequences in the nrf2 promoter.
Mol Cell Biol, 22 (2002), pp. 2883-2892
[178.]
Huang H.C., Nguyen T, Pickett CB..
Regulation of the antioxidant response element by protein kinase C-mediated phosphorylation of NF-E2-related factor 2.
Proc Natl Acad Sci U S A, 97 (2000), pp. 12475-12480
[179.]
Bloom D.A., Jaiswal AK..
Phosphorylation of Nrf2 at Ser40 by protein kinase C in response to antioxidants leads to the release of Nrf2 from INrf2, but is not required for Nrf2 stabilization/accumulation in the nucleus and transcriptional activation of antioxidant response element-mediated NAD(P)H:quinone oxidoreductase-1 gene expression.
J Biol Chem, 278 (2003), pp. 44675-44682
[180.]
Harada M, Sakisaka S, Terada K, Kimura R, Kawaguchi T, Koga H, et al.
Role of ATP7B in biliary copper excretion in a human hepatoma cell line and normal rat hepatocytes.
Gastroenterology, 118 (2000), pp. 921-928
[181.]
Kato J, Kohgo Y, Sugawara N, Katsuki S, Shintani N, Fujikawa K, et al.
Abnormal hepatic iron accumulation in LEC rats.
Jpn J Cancer Res, 84 (1993), pp. 219-222
[182.]
Sternlieb I, Quintana N, Volenberg I, Schilsky ML..
An array of mitochondrial alterations in the hepatocytes of Long-Evans Cinnamon rats.
Hepatology, 22 (1995), pp. 1782-1787
[183.]
Yamamoto H, Hirose K, Hayasaki Y, Masuda M, Kazusaka A, Fujita S..
Mechanism of enhanced lipid peroxidation in the liver of Long-Evans cinnamon (LEC) rats.
Arch Toxicol, 73 (1999), pp. 457-464
[184.]
Yamamoto H, Watanabe T, Mizuno H, Endo K, Hosokawa T, Kazusaka A, et al.
In vivo evidence for accelerated generation of hydroxyl radicals in liver of Long-Evans Cinnamon (LEC) rats with acute hepatitis.
Free Radic Biol Med, 30 (2001), pp. 547-554
[185.]
Levy E, Brunet S, Alvarez F, Seidman E, Bouchard G, Escobar E, et al.
Abnormal hepatobiliary and circulating lipid metabolism in the Long-Evans Cinnamon rat model of Wilson’s disease.
Life Sci, 80 (2007), pp. 1472-1483
[186.]
Chiba M, Itagaki S, Kobayashi M, Hirano T, Iseki K..
Characterization of hepatobiliary organic anion transporters in Long-Evans Cinnamon rats.
Drug Metab Pharmacokinet, 22 (2007), pp. 387-390
[187.]
Du C, Fujii Y, Ito M, Harada M, Moriyama E, Shimada R, et al.
Dietary polyunsaturated fatty acids suppress acute hepatitis, alter gene expression and prolong survival of female Long-Evans Cinnamon rats, a model of Wilson disease.
J Nutr Biochem, 15 (2004), pp. 273-280
[188.]
Itagaki S, Sugawara M, Kobayashi M, Miyazaki K, Hirano T, Iseki K..
Comparison of the disposition behavior of organic anions in an animal model for Wilson’s disease (Long-Evans Cinnamon rats) with that in normal Long-Evans Agouti rats.
Drug Metab Pharmacokinet, 19 (2004), pp. 150-154
[189.]
Exley C..
The pro-oxidant activity of aluminum.
Free Radic Biol Med, 36 (2004), pp. 380-387
[190.]
Bondy S.C., Guo-Ross S.X., Pien J..
Mechanisms underlying the aluminum-induced potentiation of the pro-oxidant properties of transition metals.
Neurotoxicology, 19 (1998), pp. 65-71
[191.]
Toninello A, Clari G, Mancon M, Tognon G, Zatta P..
Aluminum as an inducer of the mitochondrial permeability transition.
J Biol Inorg Chem, 5 (2000), pp. 612-623
[192.]
Klein G.L., Heyman M.B., Lee T.C., Miller N.L., Marathe G, Gourley W.K., et al.
Aluminum-associated hepatobiliary dysfunction in rats: relationships to dosage and duration of exposure.
Pediatr Res, 23 (1988), pp. 275-278
[193.]
Tanaka Y, Chen C, Maher J.M., Klaassen CD..
Kupffer cell-mediated downregulation of hepatic transporter expression in rat hepatic ischemia-reperfusion.
Transplantation, 82 (2006), pp. 258-266
[194.]
Tanaka Y, Chen C, Maher J.M., Klaassen CD..
Ischemiareperfusion of rat livers decreases liver and increases kidney multidrug resistance associated protein 2 (mrp2).
Toxicol Sci, 101 (2008), pp. 171-178
[195.]
Saris N.E., Eriksson KO..
Mitochondrial dysfunction in ischaemia-reperfusion.
Acta Anaesthesiol Scand Suppl, 107 (1995), pp. 171-176
[196.]
Morin D, Pires F, Plin C, Tillement JP..
Role of the permeability transition pore in cytochrome C release from mitochondria during ischemia-reperfusion in rat liver.
Biochem Pharmacol, 68 (2004), pp. 2065-2073
[197.]
Jaeschke H, Mitchell JR..
Mitochondria and xanthine oxidase both generate reactive oxygen species in isolated perfused rat liver after hypoxic injury.
Biochem Biophys Res Commun, 160 (1989), pp. 140-147
[198.]
Saksela M, Lapatto R, Raivio KO..
Irreversible conversion of xanthine dehydrogenase into xanthine oxidase by a mitochondrial protease.
FEBS Lett, 443 (1999), pp. 117-120
[199.]
Shibuya H, Ohkohchi N, Tsukamoto S, Satomi S..
Tumor necrosis factor-induced, superoxide-mediated neutrophil accumulation in cold ischemic/reperfused rat liver.
[200.]
Jaeschke H, Farhood A, Bautista A.P., Spolarics Z, Spitzer JJ..
Complement activates Kupffer cells and neutrophils during reperfusion after hepatic ischemia.
Am J Physiol, 264 (1993), pp. G801-G809
[201.]
Hisama N, Yamaguchi Y, Ishiko T, Miyanari N, Ichiguchi O, Goto M, et al.
Kupffer cell production of cytokine-induced neutrophil chemoattractant following ischemia/reperfusion injury in rats.
Hepatology, 24 (1996), pp. 1193-1198
[202.]
Suzuki S, Toledo-Pereyra L.H., Rodriguez F, Lopez F..
Role of Kupffer cells in neutrophil activation and infiltration following total hepatic ischemia and reperfusion.
Circ Shock, 42 (1994), pp. 204-209
[203.]
Jaeschke H..
Reactive oxygen and ischemia/reperfusion injury of the liver.
Chem Biol Interact, 79 (1991), pp. 115-136
[204.]
Jaeschke H, Bautista A.P., Spolarics Z, Spitzer JJ..
Superoxide generation by Kupffer cells and priming of neutrophils during reperfusion after hepatic ischemia.
Free Radic Res Commun, 15 (1991), pp. 277-284
[205.]
Jaeschke H, Farhood A..
Neutrophil and Kupffer cell-induced oxidant stress and ischemia-reperfusion injury in rat liver.
Am J Physiol, 260 (1991), pp. G355-62
[206.]
Zar H.A., Pretto EA..
Postischemic hepatic lipid peroxidation, monitored by chemiluminescence, depends on duration of ischemia.
Transplant Proc, 28 (1996), pp. 2980-2982
[207.]
Fukai M, Hayashi T, Yokota R, Shimamura T, Suzuki T, Taniguchi M, et al.
Lipid peroxidation during ischemia depends on ischemia time in warm ischemia and reperfusion of rat liver.
Free Radic Biol Med, 38 (2005), pp. 1372-1381
[208.]
Jaeschke H, Smith C.V., Mitchell JR..
Reactive oxygen species during ischemia-reflow injury in isolated perfused rat liver.
J Clin Invest, 81 (1988), pp. 1240-1246
[209.]
Jung S.E., Yun I.J., Youn Y.K., Lee J.E., Ha J, Noh D.Y., et al.
Effect of protease inhibitor on ischemia-reperfusion injury to rat liver.
World J Surg, 23 (1999), pp. 1027-1031
[210.]
Erdogan O, Yildiz S, Basaran A, Demirbas A, Yesilkaya A..
Effect of intraportal verapamil infusion on hepatic ischemiareperfusion injury.
Pol J Pharmacol, 53 (2001), pp. 137-141
[211.]
Bonnes-Taourel D, Guerin M.C., Torreilles J..
Is malonaldehyde a valuable indicator of lipid peroxidation?.
Biochem Pharmacol, 44 (1992), pp. 985-988
[212.]
Janero DR..
Malondialdehyde and thiobarbituric acid-reactivity as diagnostic indices of lipid peroxidation and peroxidative tissue injury.
Free Radic Biol Med, 9 (1990), pp. 515-540
[213.]
Kehrer J.P., Lund LG..
Cellular reducing equivalents and oxidative stress.
Free Radic Biol Med, 17 (1994), pp. 65-75
[214.]
Okuda M, Lee H.C., Chance B, Kumar C..
Glutathione and ischemia-reperfusion injury in the perfused rat liver.
Free Radic Biol Med, 12 (1992), pp. 271-279
[215.]
Ishii K, Arima T, Suita S..
Verapamil attenuates postischemic oxidative injury in the rat liver.
Res Exp Med (Berl), 192 (1992), pp. 151-159
[216.]
Boin I.F., Silva O.C., Souza M.E., Santos A.C., Leonardi LS..
Pyruvate kinase activation and lipoperoxidation after selective hepatic ischemia in Wistar rats.
Acta Cir Bras, 21 (2006), pp. 19-23
[217.]
Tsung A, Kaizu T, Nakao A, Shao L, Bucher B, Fink M.P., et al.
Ethyl pyruvate ameliorates liver ischemia-reperfusion injury by decreasing hepatic necrosis and apoptosis.
Transplantation, 79 (2005), pp. 196-204
[218.]
Mathews W.R., Guido D.M., Fisher M.A., Jaeschke H..
Lipid peroxidation as molecular mechanism of liver cell injury during reperfusion after ischemia.
Free Radic Biol Med, 16 (1994), pp. 763-770
[219.]
Kihara K, Ueno S, Sakoda M, Aikou T..
Effects of hyperbaric oxygen exposure on experimental hepatic ischemia reperfusion injury: relationship between its timing and neutrophil sequestration.
Liver Transpl, 11 (2005), pp. 1574-1580
[220.]
Shen S.Q., Zhang Y, Xiang J.J., Xiong CL..
Protective effect of curcumin against liver warm ischemia/reperfusion injury in rat model is associated with regulation of heat shock protein and antioxidant enzymes.
World J Gastroenterol, 13 (2007), pp. 1953-1961
[221.]
Trauner M, Fickert P, Stauber RE..
Inflammation-induced cholestasis.
J Gastroenterol Hepatol, 14 (1999), pp. 946-959
[222.]
Kubitz R, Wettstein M, Warskulat U.
Häussinger D. Regulation of the multidrug resistance protein 2 in the rat liver by lipopolysaccharide and dexamethasone.
Gastroenterology, 116 (1999), pp. 401-410
[223.]
Paulusma C.C., Kothe M.J., Bakker C.T., Bosma P.J., van B, I, van Marle J, et al.
Zonal down-regulation and redistribution of the multidrug resistance protein 2 during bile duct ligation in rat liver.
Hepatology, 31 (2000), pp. 684-693
[224.]
Almada L.L., Scandizzi A.L., Guibert E.E., Furno G, Rodriguez JV..
Biliary inorganic phosphate as a tool for assessing cold preservation-reperfusion injury: a study in the isolated perfused rat liver model.
Liver Transpl, 9 (2003), pp. 160-169
Copyright © 2008. Fundación Clínica Médica Sur, A.C.
Article options
es en pt

¿Es usted profesional sanitario apto para prescribir o dispensar medicamentos?

Are you a health professional able to prescribe or dispense drugs?

Você é um profissional de saúde habilitado a prescrever ou dispensar medicamentos