liver fibrosis in mice induced by carbon tetra chloride

Upload: sara82vet

Post on 06-Apr-2018

218 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/2/2019 Liver Fibrosis in Mice Induced by Carbon Tetra Chloride

    1/11

    Liver fibrosis in mice induced by carbon tetrachloride and its reversion by luteolin

    Robert Domitrovi a,, Hrvoje Jakovac b, Jelena Tomac c, Ivana ain d

    a Department of Chemistry and Biochemistry, School of Medicine, University of Rijeka, B. Branchetta 20, 51000 Rijeka, Croatiab Department of Physiology and Immunology, School of Medicine, University of Rijeka, Rijeka, Croatiac Department of Histology and Embriology, School of Medicine, University of Rijeka, Rijeka, Croatiad School of Medicine, University of Rijeka, Rijeka, Croatia

    a b s t r a c ta r t i c l e i n f o

    Article history:

    Received 22 June 2009Revised 26 August 2009

    Accepted 2 September 2009

    Available online 8 September 2009

    Keywords:

    Carbon tetrachloride

    Liver fibrosis

    Luteolin

    -Smooth muscle actin

    Glial fibrillary acidic protein

    Matrix metalloproteinase

    Metallothionein

    Hepatic fibrosis is effusive wound healing process in which excessive connective tissue builds up in the liver.

    Because specific treatments to stop progressive fibrosis of the liver are not available, we have investigated

    the effects of luteolin on carbon tetrachloride (CCl4)-induced hepatic fibrosis. Male Balb/C mice were treated

    with CCl4 (0.4 ml/kg) intraperitoneally (i.p.), twice a week for 6 weeks. Luteolin was administered i.p. once

    daily for next 2 weeks, in doses of 10, 25, and 50 mg/kg of body weight. The CCl 4 control group has been

    observed for spontaneous reversion of fibrosis. CCl4-intoxication increased serum aminotransferase and

    alkaline phosphatase levels and disturbed hepatic antioxidative status. Most of these parameters were

    spontaneously normalized in the CCl4 control group, although the progression of liver fibrosis was observed

    histologically. Luteolin treatment has increased hepatic matrix metalloproteinase-9 levels and metallothio-

    nein (MT) I/II expression, eliminated fibrinous deposits and restored architecture of the liver in a dose-

    dependent manner. Concomitantly, the expression of glial fibrillary acidic protein and -smooth muscle

    actin indicated deactivation of hepatic stellate cells. Our results suggest the therapeutic effects of luteolin on

    CCl4-induced liver fibrosis by promoting extracellular matrix degradation in the fibrotic liver tissue and the

    strong enhancement of hepatic regenerative capability, with MTs as a critical mediator of liver regeneration.

    2009 Elsevier Inc. All rights reserved.

    Introduction

    Liver fibrosis is a frequent event which follows a repeated or

    chronic insult of sufficient intensity to trigger a wound healing-like

    reaction, characterized by excessive connective tissue deposition in

    extracellular matrix (ECM). Chronic carbon tetrachloride (CCl4)

    intoxication is a well-known model for producing oxidative stress

    and chemical hepatic injury. Its biotransformation produces hepato-

    toxic metabolites, the highly reactive trichloromethyl free radical,

    which are further converted to the peroxytrichloromethyl radical

    (Williams and Burk, 1990). Reactive oxidant species likely contribute

    to both onset and progression of fibrosis (Poli, 2000). Antioxidant

    treatment in vivo seems to be effective in preventing or reducing

    chronic liver damage and fibrosis (Parola and Robino, 2001).

    Polyphenols, naturally occurring antioxidants in fruits, vegetables,

    andplant-derived beverages such as tea andwine, have been associated

    with a variety of beneficial properties (Havsteen, 2002). The flavone

    luteolin (3,4,5,7-tetrahydroxyflavone) is an important member of the

    flavonoid family, present in glycosylated forms and as aglycone in

    various plants (Shimoi et al., 1998). Luteolin is reported to have anti-

    inflammatory (Ziyanet al., 2007;Veda et al., 2002), antioxidant (Perez-

    Garcia et al., 2000), antiallergic (Veda et al., 2002), antitumorigenic (Ju

    et al., 2007), anxiolytic-like (Coleta et al., 2008), and vasorelaxative

    properties (Woodman and Chan, 2004). Previously, we have shown a

    hepatoprotective activity of luteolin in acute liver damage in mice

    (Domitrovi et al., 2008a, Domitrovi et al., 2009).

    Hepatic stellate cells (HSCs) are a minor cell type most commonly

    found in the space of Disse, intercalated between hepatocytes and

    cells lining the sinusoid, projecting their dendritic processes to nearby

    hepatocytes and endothelial cells (Blouin et al., 1977, Mermelstein

    et al., 2001). Upon liverinjury, HSCs become activated, converting into

    myofibroblast-like cells. Activated HSCs proliferate and produce

    extracellular matrix (ECM), playing a major role in hepatic fibrosis

    and regeneration (Friedman, 2000). ECM, which consists of collagens

    and other matrix components such as proteoglycans,fibronectins, and

    hyaluronic acid (Arthur, 1994), is regulated by a balance of synthesis

    and enzymatic degradation of ECM. The key enzymes responsible for

    degradation of all the protein components of ECM and basement

    membrane are matrix metalloproteinases (MMPs), a zinc-dependent

    family of endopeptidases. Previous studies have demonstrated that

    the activity of these enzymes is altered during the processes of

    fibrogenesis and fibrinolysis (Knittel et al., 2000).

    The metallothioneins (MTs), small cysteine-rich heavy metal-

    binding proteins, participate in an array of protectivestress responses.

    In mice, among the fourknown MTgenes, the MTI and MTII genes are

    Toxicology and Applied Pharmacology 241 (2009) 311321

    Corresponding author. Fax: +385 51 651 135.

    E-mail address: [email protected] (R. Domitrovi).

    0041-008X/$ see front matter 2009 Elsevier Inc. All rights reserved.

    doi:10.1016/j.taap.2009.09.001

    Contents lists available at ScienceDirect

    Toxicology and Applied Pharmacology

    j o u r n a l h o m e p a g e : w w w . e l s e v i e r. c o m / l o c a t e / y t a a p

    mailto:[email protected]://dx.doi.org/10.1016/j.taap.2009.09.001http://www.sciencedirect.com/science/journal/0041008Xhttp://www.sciencedirect.com/science/journal/0041008Xhttp://dx.doi.org/10.1016/j.taap.2009.09.001mailto:[email protected]
  • 8/2/2019 Liver Fibrosis in Mice Induced by Carbon Tetra Chloride

    2/11

    most widely expressed. Transcription of these genes is rapidly and

    dramatically up-regulated in response to agents which cause

    oxidative stress and/or inflammation (Andrews, 2000). The induction

    of MT synthesis can protect animals from hepatotoxicity induced by

    various toxins including CCl4, but also play a role in repair and

    regeneration of injured liver (Cherian and Kang, 2006).

    Because the specific treatments to stop progressive fibrosis of the

    liver are not available, the objective of the present study was to

    investigate the therapeutic effect and mechanisms of action of luteolinin chemically induced liver fibrosis in mice.

    Materials and methods

    Materials. Luteolin, carbon tetrachloride (CCl4), olive oil, dimethyl

    sulfoxide (DMSO), nitric acid (HNO3), hydrogen peroxide (H2O2), and

    gelatin were obtained from Sigma Chemical Co. (St. Louis, MO, USA).

    All other chemicals and solvents were of the highest grade commer-

    cially available.

    Animals. Male Balb/c mice from our breeding colony, 23 months

    old, were divided into 6 groups with 5 animals per group. Mice were

    fed a standard rodent diet (pellet, type 4RF21 GLP, Mucedola, Italy)

    containing 19.4% protein, 5.5% fiber, 11.1% water, 54.6% carbo-

    hydrates, 6.7% ash, and 2.6% by weight of lipids (native soya oil) to

    prevent essential fatty acid deficiency. Total energy of the diet was

    16.4 MJ/kg. The animals were maintained at 12 h light/dark cycle, at

    constant temperature (201 C) and humidity (505%). All

    experimental procedures were approved by the Ethical Committee

    of the Medical Faculty, University of Rijeka.

    Experimental design. Mice were given CCl4 intraperitoneally (i.p.) at

    a dose of 0.4 ml/kg, dissolved in olive oil, twice a week for 6 weeks

    (the CCl4 group), except the control group which received vehicle

    only. Seventy-two hours after the last CCl4 injection, the CCl4 group

    was killed. The CCl4 control group was observed for spontaneous

    resolution of hepatic fibrosis for next 2 weeks. In the luteolin-treated

    groups, the polyphenolic compound was administered i.p. at a dose of

    10, 25, or 50 mg/kg daily for 2 weeks, respectively. These doses wereselected on the basis of preliminary studies (Domitrovi et al., 2008a,

    Domitrovi et al., 2009). Luteolin was dissolved in DMSO and diluted

    in saline to the final concentrations. The final concentration of DMSO

    was less than 1%. Mice from the control and CCl4 control groups

    received diluted DMSO solution daily for 2 weeks. Animals were

    terminated 24 h after the last dose of luteolin or diluted solvent by

    cervical dislocation. The blood was taken from orbital sinus of ether

    anesthetized mice. The abdomen of terminated animal was cut open

    quickly and the liver perfused with isotonic saline, excised, blotted

    dry, weighed, and divided into samples. The samples were used to

    assess biochemical parameters, and another was preserved in a 4%

    phosphate-buffered formalin solution to obtain histological sections.

    Hepatotoxicity study. Serum levels of ALT, AST, and ALP as markersof hepatic function, were measured by using a Bio-Tek EL808 Ultra

    Microplate Reader (BioTek Instruments, Winooski, VT, USA) according

    to the manufacturer's instructions.

    Determination of Cu/Zn SOD activity and GSH concentration. Cu/Zn

    SOD activity and total GSH, indicators of oxidative stress, were

    measured spectrophotometrically, using Superoxide Dismutase Assay

    Kit andGlutathioneAssayKit (Cayman Chemical,Ann Arbor,MI, USA),

    according to the manufacturer's instructions. Protein content in

    supernatants was estimated by Bradford's method, with bovine

    serum albumin used as a standard (Bradford, 1976).

    Determination of hepatic hydroxyproline. The tissue samples

    (50 mg) were hydrolyzed in 4 ml 6 M HCl at 110 C for 24 h. After

    being filtered through a 0.45-m filter, 2 ml of samples was extracted

    and analyzed according to the procedure of Bergman and Loxley

    (1963). Briefly, sample neutralization was obtained with 10 M NaOH

    and 3 M HCl. After neutralization, subsequent steps were made in

    duplicate for each sample. To a 200 l of the above solution, 400 l of

    isopropanol in citrate-acetate-buffered Chloramine T was added. After

    4 min, 2.5 ml of Ehrlich reagent was added. Tubes were wrapped in

    aluminum foil and incubated for 25 min in a water-bath at 60 C,

    cooling each sample in tap water, and measuring the absorbance ofeach sample spectrophotometrically at 550 nm (Cary 100, Varian,

    Mulgrave, Australia).

    Determination of trace elements. Hepatic zinc (Zn) and copper (Cu)

    content were determined by ion coupled plasma spectrometry (ICPS)

    using Prodigy ICP Spectrometer (Leeman Labs, Hudson, NH, USA),

    accordingto the method previouslydescribed (Domitroviet al., 2008a).

    Determination of retinol. The hepatic levels of retinol were analyzed

    by high-performance liquid chromatography (HPLC) according to

    Hosotani and Kitagawa (2003), as described previously (Domitrovi

    et al., 2008b).

    Histopathology. Liver specimens were fixed in 4% phosphate-

    buffered formalin, embedded in paraffin, and cut into 4 m thick

    sections. Sections for histopathological examination were stained

    with hematoxylin and eosin (H&E) and Mallory trichrome stain using

    standard procedure.

    Immunohistochemical determination of GFAP, -SMA, and MT I/II.

    Immunohistochemical studies were performed on paraffin embedded

    liver tissues using mouse monoclonal anti-MT I+II antibody diluted

    1:50 (clone E9; DakoCytomation, Carpinteria, CA, USA), mouse

    monoclonal anti-GFAP antibody diluted 1:100 (clone 1B4; BD

    Pharmingen, San Diego, CA, USA), and mouse monoclonal antibody

    to -SMA diluted 1:100 (SPM332; Abcam, Cambridge, UK),

    employing DAKO EnVision+ System, Peroxidase/DAB kit according

    to the manufacturer's instructions (DAKO Corporation, Carpinteria,

    CA, USA). Briefly, slides were incubated with peroxidase block toeliminate endogenous peroxidase activity. After washing, monoclonal

    antibodies diluted in phosphate-buffered saline supplemented with

    bovine serum albumin were added to tissue samples and incubated

    overnight at 4 C in a humid environment, followed by incubation

    with peroxidase labeled polymer conjugated to secondary antibodies

    containing carrier protein linked to Fc fragments to prevent

    nonspecific binding. The immunoreaction product was visualized by

    adding substrate-chromogen diaminobenzidine (DAB) solution,

    resulting with brownish coloration at antigen sites. Tissues were

    counterstained with hematoxylin, dehydrated in gradient of alcohol,

    and mounted with mounting medium. The intensity of staining was

    graded as weak, moderate, and intense. The specificity of the reaction

    was confirmed by substitution of primary antibodies with irrelevant

    immunoglobulins of matched isotype, used in the same conditionsand dilutions as primary antibodies. Stained slides were analyzed by

    light microscopy (Olympus BX51, Tokyo, Japan).

    MMP zymography. MMP-2 and MMP-9 activities were analyzed by

    gelatin zymography assays as described (Kuo et al., 2003), with

    modifications. After tissue homogenization in radioimmuno-

    precipitation assay buffer (4 ml of buffer per gram of tissue)

    containing 50 mM TrisHCl pH 7.4, 150 mM NaCl, 1% NP-40, 0.5%

    sodium deoxycholate, 0.1% SDS, 2 mM PMSF, 1 mM sodium

    orthovanadate, and 2 g/ml of each aprotinin, leupeptin and

    pepstatin. 10 g of liver tissue protein lysates were separated by a

    10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-

    PAGE) gels containing0.1%gelatin, at 4 C and 150 V for 4 h. Gels were

    washed for 30 min in 2.5%Triton X-100 to remove the SDS, and briefly

    312 R. Domitrovi et al. / Toxicology and Applied Pharmacology 241 (2009) 311321

  • 8/2/2019 Liver Fibrosis in Mice Induced by Carbon Tetra Chloride

    3/11

    washed in the reaction buffer containing 50 mM TrisHCl, pH 7.5,

    5 mM CaCl2, 1 M ZnCl2, and 0.02% NaN3. The reaction buffer was

    changed to a fresh one, and the gels were incubated at 37 C for 48 h.

    Gelatinolytic activity was visualized by staining the gels with 0.1%

    Coomassie blue R-350, destained with methanol-acetic acid water

    (30:10:60 v/v) two times for 20 min. The clear zones in the

    background of blue staining indicate the presence of gelatinase

    activities. The intensity of the bands was assayed by scanning video

    densitometry (Bio-Rad GS-710 Calibrated Imaging Densitometer, Bio-

    Rad Laboratories, UK).

    Statistical analysis. Data were analyzed using StatSoft STATISTICA

    version 7.1 software. Differences between the groups were assessed

    by a nonparametric ManWhitney and KruskalWallistests. Values in

    the text are means standard deviation (SD). Differences with

    pb0.05 were considered to be statistically significant.

    Results

    Hepatotoxicity

    Serum AST, ALT, and ALP activities were changed significantly in

    mice receiving CCl4 twice a week for 6 weeks and terminated 72 h

    later (Table 1). The relative liver weight significantly increased in the

    CCl4 group when compared to controls. In the CCl4 control group,

    observed for the spontaneous regression offibrosis for 2 weeks, therelative liver weight and ALP activity were still increased; however,

    AST and ALT activities were normalized. Luteolin administration

    attenuated the elevation of ALP activity in the mice treated with CCl 4and decreased relative liver weight to control values. Higher doses of

    luteolin, 25 and 50 mg/kg, decreased ALP activity below control

    values. However, AST activity in mice treated with 50 mg/kg of

    luteolin was significantly increased compared to the control group

    and groups treated with luteolin.

    Hepatic hydroxyproline

    The liver hydroxyproline content was fivefold higher in the CCl4group than in controls and progressively increased in the CCl4 control

    group (Table 1). Luteolin therapy significantly decreased the hepatichydroxyproline level, which was returned to normal values in the

    group receiving 50 mg/kg of luteolin.

    Cu/Zn SOD activity and GSH concentration

    There was no evidence of oxidative stressin the CCl4 control group

    when compared to the CCl4 group. The hepatic total GSH level

    remained at normal values throughout the treatment period, except

    in the group treated with 50 mg/kg of luteolin, where it was

    significantly increased (Table 2). Cu/Zn SOD activity was increased in

    the CCl4 group; however, its level was normalized in the CCl4 control

    group and groups treated with luteolin. The liver total retinol

    concentration was markedly decreased in the CCl4 group, then

    increased in the CCl4 control group, returning to normal values in

    the group treated with the highest dose of luteolin.

    Cu and Zn content

    The Cu content was decreased in all experimental groups,

    compared to controls (Table 2). The hepatic Zn content has not

    been significantly changed by CCl4 intoxication or during luteolin

    therapy. The liver trace element content was expressed as g/g dry

    liver weight (ppm).

    Histopathology

    Liver sections from control mice stained with hematoxylin and

    eosin (H&E) showed normal hepaticarchitecture.Liver specimensfrom

    the CCl4 group showed remnants of degenerated and balooned/necrotic hepatocytes containing acidophilic hyaline inclusions

    (Fig. 1B). Mild mononuclear cell infiltration in these areas was also

    present. Hepatocytes in the vicinity of hepatic lesions, particularly

    those situated immediately alongside the lesion border, showed

    features of pronounced macrovesicular or microvesicular steatosis.

    Distendedsinusoidal spacesfilled witherythrocytes as a morphological

    hallmark of congestion (disturbed blood flow) were also observed.

    Hepatic lesions, fatty degeneration, and hyaline deposits as well as

    morphological signatures of congestion were more pronounced in the

    CCl4 control group (Fig. 1C). Hepatic lesions and hyaline deposits were

    also present in the livers of mice treated with 10 mg/kg of luteolin

    (Fig. 1D), but they were significantly reduced in extent and were less

    frequent compared to the CCl4 control group, whereas macrovesicular

    steatosis was still present in the surrounding liver parenchyma, withnoticeable signs of congestion. The livers of mice receiving 25 mg/kg of

    luteolin showed only sporadic, markedly small hepatic lesions, hyaline

    Table 1

    Relative liver weight, hydroxyproline content, and serum markers of liver damage.

    Control CCl4 CCl4 control Luteolin 10 mg/kg Luteolin 25 mg/kg Luteolin 50 mg/kg

    Relative liver weight (g/100 g body weight) 5.31 0.40a 6.940.23b 6.170.19b 5.830.65a 5.340.20a 4.950.72a

    Hydroxyproline (g/g liver) 189 37a 846159 b 1188205c 44382d 29434e 23131a

    AST (U/l) 22.7 2.5a 276.54.1b 20.41.6a 18.63.3a 23.34.9a 27.010.3a

    ALT (U/l) 16.0 3.7a 226.120.2b 13.12.1a 15.12.9a 14.02.3a 12.01.2a

    ALP (U/l) 77.4 1.1a 114.04.0b 87.97.9b 79.84.1a 66.73.4c 60.35.0c

    Each value represents the meanSD for 5 mice. Values not sharing a common letter superscript (a, b, c) are significantly different (pb0.05). Statistical difference was determined

    using nonparametric ManWhitney and KruskalWallis tests.

    Table 2

    Hepatic indicators of oxidative stress, liver retinol, and metal content.

    Control CCl4 CCl4 contro l Luteo lin 10 mg/kg Luteolin 25 mg/kg Luteolin 50 mg/kg

    Cu/Zn SOD (U/mg protein) 0.950.04a 1.500.10b 0.910.23a 0.860.08a 0.880.08a 0.890.15a

    GSH (M/g liver) 9.71 0.51a 9.640.21a 9.510.63a 9.560.79a 10.570.60a 11.370.65b

    Retinol (M/g liver) 1.51 0.13a 0.740.09b 1.220.11c 1.020.19c 1.190.12c 1.320.23a,c

    Zn (ppm) 75.3 2.6 85.3 9.2 84.8 10.4 79.3 6.1 81.3 9.1 85.3 11.6

    Cu (ppm) 22.5 1.7a 14.01.3b 16.72.1b,c 16.30.7c 16.10.8c 15.80.4c

    Each value represents the meanSD for 5 mice. Values not sharing a common letter superscript (a, b, c) are significantly different (pb0.05). Statistical difference was determined

    using nonparametric Man

    Whitney and Kruskal

    Wallis tests.

    313R. Domitrovi et al. / Toxicology and Applied Pharmacology 241 (2009) 311321

  • 8/2/2019 Liver Fibrosis in Mice Induced by Carbon Tetra Chloride

    4/11

    changes, and microvesicular steatosis in the periportal areas, but

    without morphological signs of hemodynamic disturbance (Fig. 1E). In

    animals treated with 50 mg/kg of luteolin, the livers showed

    maintained histoarchitecture, almost similar to controls, with only

    weak microvesicular steatosis of periportal and perilobular hepato-

    cytes (Fig. 1F). There were no changes which could be attributed to

    hepatic hemodynamic disturbances in these animals.

    The livers from control mice stained with Mallory trichrome stain

    showed traces of collagen only in the vascular walls. Liver section from

    the CCl4 group showed multiple fibrotic nodules and extensive fibrosis

    predominantly in the periportal areas (Fig. 2B). The pericellular

    collagen deposition and the extent of fibrotic changes were even

    more pronounced in the CCl4 control group (Fig. 2C). Fibrotic lesions

    were still present in thelivers of micetreated with 10 mg/kg of luteolin

    (Fig. 2D), but they were significantly reduced in extent and were less

    frequent compared to the CCl4 control group, whereas mild pericellular

    fibrosis wasobserved in thesurrounding liver parenchyma. Thelivers of

    mice receiving 25 mg/kg of luteolin showed only sporadic, markedly

    Fig. 1. The effect of CCl4 and luteolin therapy on liver histology. (A) The livers from control mice showed normal hepatic architecture. (B) The livers of mice receiving CCl4 twice a

    week for 6 weeks, terminated 72 h after the last CCl4 injection, showed severe hepatic lesions, degenerated and balooned/necrotic hepatocytes with cellular hyaline inclusions, and

    mild mononuclear cell infiltration (arrowheads). (C) In the CCl4 control group, observed for spontaneous regression offibrosis for additional 2 weeks, hepatic lesions, balooned

    hepatocytes, and hyaline bodies were more frequent. (D) Hepatic lesions and hyaline deposits in mice receiving 10 mg/kg luteolin for 2 weeks were signi ficantly reduced in extent

    and were less frequent. (E) The livers of mice receiving 25 mg/kg of luteolin showed sporadic, small hepatic lesions, hyaline changes, and microvesicular steatosis. (F) Mice treated

    with 50 mg/kg of luteolin showed maintained hepatic architecture, with only weak microvesicular steatosis. Arrows show balooned hepatocytes with hyaline inclusions. Original

    magnification 100, insets 1000. H&E stain.

    314 R. Domitrovi et al. / Toxicology and Applied Pharmacology 241 (2009) 311321

  • 8/2/2019 Liver Fibrosis in Mice Induced by Carbon Tetra Chloride

    5/11

    small fibrotic deposits in the periportal areas, with a discrete peri-

    cellular fibrosis (Fig. 2E). Animals treated with 50 mg/kg of luteolin

    showed only minor and sporadic pericellularfibrotic changes (Fig. 2F).

    Immunohistochemistry of GFAP, -SMA, and MT I/II

    Activated HSCs could be distinguished from the other myofibro-

    blasts of the liver (such as portal myofibroblasts) by their specific

    positionin the liver parenchyma (Cassiman et al.,2002). With regard tothe distribution of -SMA-positive fibrogenic cells, in the livers of

    controlanimals,-SMA immunopositivity was restrictedto the smooth

    musculature belongingto thearterial tunica media, as well as to thewall

    of majority of portal and central veins, while other liver cells remain

    negative (Fig. 3A). CCl4 strongly induced perisinusoidal -SMA

    expression, which was recognized as activated HSCs, through affected

    lobuli, connected between themselves with thin, bridging immuno-

    positivity (Fig. 3B). The HSCsvaried inshape and size. In the CCl4 control

    group, perisinusoidal immunopositivity was withdrawn, but emerged

    in hepatocytes located at the scar-parenchyma interface (Fig. 3C). Thelivers of mice receiving 10 and 25 mg/kg of luteolin showed only

    Fig. 2. The histopathologic detection of collagen in the livers. Collagen fibers of theconnective tissues areidentified by their blue color.(A) Liver section from a control mice. (B)Mice

    receiving CCl4 twice a week for6 weeks,terminated 72h after thelast CCl4 injection, had developed extensivefibrosisin theperiportal areas.(C) TheCCl4 control group, observed for

    spontaneousregression offibrosisfor additional 2 weeks,had more intense collagen staining.(D) Thecollagendeposits in mice receiving10 mg/kg luteolin for2 weeks were greatly

    reduced in size. (E)The livers of mice receiving25 mg/kg of luteolin showedsporadic, smallfibroticlesions in theperiportal zone. (F)In mice treated with 50mg/kgof luteolin there

    were observed only minor pericellular fibrotic changes (arrow). Original magnification 100, insets 1000. Mallory trichrome stain.

    315R. Domitrovi et al. / Toxicology and Applied Pharmacology 241 (2009) 311321

  • 8/2/2019 Liver Fibrosis in Mice Induced by Carbon Tetra Chloride

    6/11

    sporadic -SMA immunopositivity in the hepatocytes in the vicinity offibroticlesions (Figs. 3D and E). The livers ofmicereceiving50 mg/kg of

    luteolin showed staining pattern similar to control animals.

    Immunohistochemical staining using monoclonal anti GFAP anti-

    body showed thin, irregular immunopositive bands lining the hepatic

    sinusoids of control animals (staining pattern considered as a

    characteristic for HSC) (Fig. 4A). In the CCl4 and CCl4 control groups,

    moderate and strong GFAP expressions, respectively, were limited to

    the scar areas (Figs. 4B and C). Treatment with 10 and 25 mg/kg ofluteolin decreased GFAP expression in the fibrotic areas, but induced

    sporadic assembling of GFAP positive cells in the surrounding tissue

    (Figs. 4D and E). The livers of mice receiving 50 mg/kg of luteolin

    showed staining pattern similar to control animals, characterized by

    moderate GFAP expression alongside the sinusoidal border (Fig. 4F).

    Control animals had moderate cytoplasmic MT I/II expression in

    the perilobular areas (Fig. 5A). Liver tissues from the CCl4 group

    showed weak cytoplasmic immunostaining of fatty changed hepa-

    tocytes surrounding the fibrotic areas, but strong immunopositivity

    within balooned hepatocytes (Fig. 5B). In the CCl4 control group, MTI/II immunopositivity was found to be only sporadicand restricted to

    Fig. 3. Theexpression andspecific tissue distributionof-SMA. (A)In thelivers of control animals,-SMAimmunopositivitywas restricted to smooth musculature(arrow), whileother

    livercells remain negative.(B) In micereceiving CCl4 twicea week for6 weeks,terminated72 h afterthe last CCl4 injection,many-SMA-containingmyofibroblastswere strongly and

    diffuselystainedin affectedlobuli, connectedbetweenthemselves withthin, bridging immunopositivity (arrows).(C) In the CCl4 control group, observed for spontaneous regression

    offibrosis foradditional2 weeks, clustersof moderately-SMA-immunopositive hepatocytes locatedat thescar-parenchymainterfacewere found(arrows).The livers ofmice receiving

    10 mg/kg (D) and 25 mg/kg (E) of luteolin showed only sporadic -SMA immunopositivity in the hepatocytes in vicinity to fibrotic lesions (arrows). (F) The livers of mice receiving

    50 mg/kg of luteolin showed staining pattern similar to control animals. Original magnification 400, inset 1000. Immunohistochemical stain for -SMA.

    316 R. Domitrovi et al. / Toxicology and Applied Pharmacology 241 (2009) 311321

  • 8/2/2019 Liver Fibrosis in Mice Induced by Carbon Tetra Chloride

    7/11

    hyaline bodies in the context offibrous lesions, while in the remnant

    liver tissuewas not detected(Fig. 5C). The livers of animals receiving

    10 mg/kg of luteolin showed weak cytoplasmic MT I/II expression in

    the remaining hepatic parenchyma and more frequent appearance of

    MT I/II immunopositive hyaline deposits (Fig. 5D). Concomitantly

    with the histoarchitecture improvement, moderate perilobular

    MT I/II immunopositivity was observed in the livers of animals

    treated with 25 mg/kg of luteolin (Fig. 5E). Liver sections from mice

    treated with 50 mg/kg of luteolin have shown strong cytoplasmic and

    nuclear immunopositivity, remarkably in the periportal and perilob-

    ular areas (Fig. 5F). Fig. 5G shows the extent of MT I/II expression.

    MMP zymography

    MMPs were identified by their molecular weights. Liver extracts

    contained mainly the latent form of MMP-9 at about 92 kDa (Fig. 6).

    CCl4-intoxication decreased MMP-9 expression, which was even more

    pronounced in the CCl4 control group. Treatment with lutolin resulted

    in an increasein MMP-9 expressionin a dose-dependentmanner, being

    the most prominent in mice treated with 25 and 50 mg/kg of luteolin.

    Active form of MMP-9 was barely detectable in groups treated with 25

    and 50 mg/kg of luteolin. In contrast to MMP-9, both latent and active

    forms of MMP-2 expression were below detection limit in all groups.

    Fig. 4. The expression and specific tissue distribution of GFAP. (A) The liver sections from control mice showed thin GFAP immunopositive bands lining the hepatic sinusoids. (B) In

    the livers of mice receiving CCl4 twice a week for 6 weeks, terminated 72 h after the last CCl4 injection, moderate GFAP expression, and (C) in mice from the CCl4 control group,

    observed for spontaneous regression offibrosisfor additional 2 weeks,strong GFAP expression were limited to thescarareas. Mice receiving10 mg/kg (D)and (E)25 mg/kg luteolinfor 2 weeks had decreased GFAP expression in the fibrotic areas, with sporadic expression in the surrounding tissue. (F) The livers of mice receiving 50 mg/kg of luteolin showed

    staining pattern similar to the control animals. Original magnification 1000. Immunohistochemical stain for GFAP.

    317R. Domitrovi et al. / Toxicology and Applied Pharmacology 241 (2009) 311321

  • 8/2/2019 Liver Fibrosis in Mice Induced by Carbon Tetra Chloride

    8/11

    318 R. Domitrovi et al. / Toxicology and Applied Pharmacology 241 (2009) 311321

  • 8/2/2019 Liver Fibrosis in Mice Induced by Carbon Tetra Chloride

    9/11

    Discussion

    Liver fibrosis represents chronic wound repair following liverinjury (Friedman, 1999). Independently from the nature of the injury,

    the usual initiating event of fibrogenesis is represented by tissue

    necrosis of variable entity. Activation of either resident or blood-

    derived phagocytes increases the steady-state concentration of

    fibrogenic cytokines, which recruit a large amount offibroblasts and

    fibroblast-like cells for excess production of ECM. The terminal

    outcome of liver fibrosis is the formation of nodules encapsulated

    by fibrillar scar matrix (Tsukamoto, 1999). Irreversible fibrosis

    develops when the appropriate cellular mediators as the source of

    MMPs are absent (Issa et al., 2004). The progression of fibrosis

    observed in the CCl4 control group suggests development of an

    irreversible fibrosis. Additionally, numerous eosinophilic hyaline

    globules of varying sizes have been found in fibrotic lesions. The

    amount of hyaline bodies has correlated with abundance of collagendeposits and the degree of liver fibrosis. Hyaline deposits, such as

    Mallory bodies, could be occasionally found in the hepatocytes in

    alcoholic hepatitis but also in different nonalcoholic liver disorders

    (Denk et al., 2000). These inclusions consist of aggregates of

    abnormally phosphorylated, ubiquitinated, and cross-linked keratins

    and nonkeratin components (Stumptner et al., 2002). Protein

    aggregates could be reverted to normalstate by molecularchaperones

    or degradated by proteasomes (Kopito, 2000). However, if reparation

    or degradation fails, abnormal proteins became segregated in the

    cytoplasm as inclusion bodies.

    Hepatic indicators of oxidative stress and hepatic damage in this

    study were markedly changed in CCl4-intoxicated group, 72 h after

    discontinuation of the toxin. Two weeks later, in the CCl4 control

    group and the groups treated with luteolin, most of the biochemical

    parameters, Cu/Zn SOD activity, GSH, and aminotransferase levels did

    not indicate signifi

    cant oxidative stress or hepatocellular damage. Thepossible reasonfor this is that mammalian cells respond quickly to the

    initial oxidative damage by means of a characteristic adaptative

    phenomenon (Radice et al., 1998). Acquisition of adaptation to tissue

    damage is a common response to insults in order to reduce the

    vulnerability of the cell or tissue, which depends on the activity of

    various stress sensors, signal transduction and effectors of protection

    (Hagberg et al., 2004). Therefore, biochemical parameters may not

    reflect the degree of tissue damage (Murayama et al., 2007). These

    results are of the significant clinical importance, suggesting that the

    classical biochemical parameters such as serum aminotransferase

    activities are not a reliable indicators of chronic liver damage.

    The accumulation of ECM observed in fibrosis and cirrhosis is

    considered to be the result of activation offibroblasts, which acquire a

    myofibroblastic phenotype. Myofibroblasts are produced by the

    activation of precursor cells, such as HSCs and portal fibroblasts.

    Significant differences between these two fibrogenic cell populations

    have been reported, in the mechanisms leading to myofibroblast

    differentiation, their activation and deactivation (Guyot et al., 2006).

    However, the pathophysiological mechanisms of hepatic fibrogenesis

    are not yet fully understood. In particular, the role of HSCs remains

    unclear. HSCs have been also found to migrate in vitro (Marra et al.,

    1999), suggesting that they may migrate to the fibrotic lesions and

    take part in the repair process (Kinnman et al., 2000). Stimulation of

    HSCs with various cytokinesor type I collagen,results in an increase in

    their migratory capacity, which could be accompanied by or

    independent of HCS proliferation (Yang et al., 2003). In the quiescent

    state, HSCs express desmin and GFAP (Yokoi et al., 1984, Neubauer

    et al., 1996). Upon HSCs activation, GFAP expression decreases,

    whereas the levels of desmin and -SMA increase, due to theirtransformation into myofibroblast-like cells (Campbell et al., 2005).In

    this study, deposition of collagen in damaged hepatic areas, associated

    with increased -SMA and decreased GFAP perisinusoidal immuno-

    positivity, indicatesthat activated HSCs are responsible for thefibrosis

    seen in the CCl4-intoxicated mice. Additionally, increased GFAP

    immunopositivity in fibrotic scars suggests the accumulation of

    quiescent HSCs at the places of active fibrogenesis. Changes in the

    activation marker expression in the CCl4 control group, however,

    indicate deactivationof HSCs, which is critical for resolution offibrosis

    (Iredale, 2001). Nevertheless, -SMA immunopositive hepatocytes

    located at the scar-parenchyma interface in the CCl4 control group

    suggest that hepatocytes have acquired myofibroblast-like pheno-

    type. Indeed, Zeisberg et al. (2007) have recently shown that adult

    hepatocytes could be actively engaged in the epithelial to mesenchy-mal transition mediated by TGF-1, by expressing fibroblast-specific

    protein-1 (FSP1), suggesting that hepatocyte-derived fibroblasts

    could be an additional lineage of mesenchymal cells that contribute

    to the progression of liver fibrosis. Furthermore, Dooley et al. (2008)

    have observed hepatocyte transdifferentiation immediately adjacent

    to the fibrotic lesions, which is in agreement to ourfindings. However,

    to our knowledge, this is for the first time that the -SMA expression,

    Fig. 6. A representative SDS-PAGE gelatin zymogram of liver specimens from thecontrol and treated animals. (A) Expression of latent form of 92 kDa MMP-9 in equal

    protein quantities of hepatic homogenates from control mice (lane 1), mice treated

    with CCl4 (lane 2), mice left for spontaneous recovery (lane 3), and from mice treated

    with luteolin 10 mg/kg (lane 4), 25 mg/kg (lane 5), and 50 mg/kg (lane 6). Weak

    expression of theactive 86 kDaform ofMMP-9was noticeable onlyin lanes 5 and6. No

    significant effect of CCl4-intoxication or the treatment with luteolin on the MMP-2

    expression or proMMP-2 activation has been detected. (B) Densitometric analysis of

    bands detected in gelatin zymograms. The bar graph shows averages of band intensity

    of proMMP-9 expression (meanSD, n =5). Values not sharing a common letter

    superscript (a, b, c, d, e) are significantly different (pb0.05). MMPs were identified by

    their molecular weights.

    Fig. 5. The expression and specific tissue distribution of MT I/II. (A) The livers from control mice showed weak perilobular MT I/II immunopositivity. (B) The livers of mice receiving

    CCl4 twice a week for6 weeks,terminated 72 h after thelast CCl4 injection, showedweak cytoplasmicimmunostainingof hepatocytes located at the scar-parenchymainterface, with

    strong immunopositivity in the fibrotic lesions. (C) In the CCl4 control group, observed for spontaneous regression offibrosis for additional 2 weeks, immunopositivity was only

    sporadic and restricted to the fibrotic scars. (D) Mice receiving 10 mg/kg luteolin therapy for 2 weeks had weak cytoplasmic MT I/II expression with strong immunopositivity in the

    fibroticlesions.(E) Thelivers ofmice receiving25 mg/kg of luteolin showedmoderateperilobularMT I/II immunopositivity. (F)Strong both cytoplasmicand nuclear perilobular and

    periportal staining in the group treated with 50 mg/kg of luteolin. (G) The bar graph shows the extent of hepatocellular MT I/II expression (meanSD, n =5). Values not sharing a

    common letter superscript (a, b, c, d, e) are significantly different (pb

    0.05). Original magnification 100. Immunohistochemical stain for MT I/II.

    319R. Domitrovi et al. / Toxicology and Applied Pharmacology 241 (2009) 311321

  • 8/2/2019 Liver Fibrosis in Mice Induced by Carbon Tetra Chloride

    10/11

    characteristic for activated HSCs, is found in hepatocytes. This

    subpopulation of hepatocytes could contribute to collagen deposition

    observed in the CCl4 control group. Perisinusoidal GFAP expression in

    the group treated with 50 mg/kg of luteolin indicates a complete

    return of HSCs into a quiescent state.

    Morphological studies have shown that fibrotic lesion of the liver

    also typically involves, beside an accumulation of extracellular

    collagen matrix and a transformation of stellate cells to actively

    dividing myofi

    broblasts, a reduction in stellate cell lipid droplets andhepatic storage of total retinol (Blomhoff and Wake, 1991). Decreased

    hepatic total retinol level in the CCl4 group observed in this study is in

    agreement with previous studies (Seifert et al., 1994, Natarajan et al.,

    2005). The vitamin A status of the liver plays an important role in

    hepatic fibrogenesis. The reduction of retinoid levels in HSC by an

    enhanced secretionof retinol from theliver into thecirculationduring

    CCl4 treatment may stimulate the transformation of these cells intofibroblasts and contribute to fibrogenesis (Seifert et al., 1994).

    Increased liver retinol observed in the CCl4 control group, as well as

    in mice receiving luteolin therapy, also indicates a decrease in the

    number of activated HSCs.

    In normal tissues, matrix protein degradation is accomplished by a

    family of MMPs, whose expression significantly increases in the case

    of acute liver injury (Arthur, 1994). In chronic liver injury sustained

    tissue damage and inflammation also induce MMPs overexpression

    (Kossakowska et al., 1998). MMP-2, the 72-kD collagenase IV/

    gelatinase A, is an important factor in the metabolism of type IV

    collagen in sinusoid basement, produced by HSCs. MMP-9, the 92-kD

    collagenase IV/gelatinase B, a major MMP in basement membrane-

    like ECM remodeling, is secreted by neutrophil granulocytes, macro-

    phages and leukocytes as a major source for MMP-9 expression

    (Salguero Palacios et al., 2008), with HSCs as an additional source

    (Knittel et al., 1999). MMP activity decreases with the progression of

    liver fibrosis (Iredale, 2001). Liver injury which results in HSC

    activation, particularly if chronic, leads to an increase in overall

    numbers of myofibroblast-like activated HSCs that are actively

    producing matrix, while simultaneously preventing degradation of

    the matrix through expression of TIMPs. It has been consensually

    accepted that increased production and activity of TIMPs are requiredfor decrease in MMP activity (Perez-Tamayo et al., 1987, Benyon and

    Iredale, 2000). The key events in the reversion of liver fibrosis include

    apoptosis of HSCs, increased MMP activity, decreased expression of

    TIMPs, and increased degradation of collagen (Woessner, 1991). In

    the liver, myofibroblasts derived from hepatic stellate cells undergo

    apoptosis during the spontaneous resolution of liver fibrosis induced

    by CCl4 treatment (Iredale et al., 1998). During spontaneous recovery

    from liver fibrosis, TIMP-1 level decreases, and collagenase activity

    and the apoptosis of hepatic stellate cells increase, which results in an

    increasedfibrolysis and removal of excessively deposited ECM. MMPs

    could be derived either from activated HSCs prior to or during

    apoptosis, from quiescent HSCs, or from other liver cells (Iredale,

    2001). It is still unknown how the fibrotic tissues are removed in vivo

    and how MMPs, which are usually down-regulated in establishedfibrotic tissues, are expressed again during the resolution process

    (Han, 2006). Our result showed that CCl4-intoxication decreased

    MMP-9 expression in CCl4 group 72 h after toxin discontinuation, with

    further decrease in the CCl4 control group 2 weeks later. Luteolin

    therapy has induced the expression of MMP-9, with the highest

    increase in the group treated with 50 mg/kg. The MMP-9 production

    by other cells distinct from HSCs could explain its increased

    expression despite HSC deactivation. In contrast, MMP-2 did not

    play a significant role in this model, which was confirmed by

    immunohistochemistry (data not shown).

    The mechanisms by which MT participates in the pathology and

    the reversion of liver fibrosis are still largely unknown. Although a

    number of biological functions have been proposed for MTs, most of

    them are related to its metal-binding property, mainly Zn and Cu

    homeostasis and heavy metal detoxification (Andrews, 2000).

    Recently, we have shown dose- and time-dependent up-regulation

    of MTs expression by luteolin in acute hepatotoxicity induced by CCl4and the enhancement of hepatic regenerative capability (Domitrovi

    et al., 2008a). Hepatic MT I/II expression in necrotic areas was

    correlated with the Zn and Cu content. However, MTs expression in

    chronic exposure to CCl4 in this study was not correlated with either

    the Zn or Cu hepatic content. Increased MT synthesis also occurs in

    response to oxidative stress (Chubatsu and Meneghini, 1993). In thisstudy, disturbance in antioxidative status in the CCl4 control groupand

    groupstreated with luteolin was not observed, therefore we could not

    explain the down-regulation of MT I/II as a consequence of oxidative

    stress. Furthermore, MT has the essential role in cell proliferation and

    liver regeneration (Apostolova and Cherian, 2000). Immunohisto-

    chemical studies have demonstrated increased MT expression both in

    the cytoplasm and nucleus of rapidly proliferating cells (Miles et al.,

    2000). MT may donate Zn and Cu to certain metalloenzymes and

    transcription factors associated with gene transcription, cell replica-

    tion, differentiation, and growth (Wostowski, 1993, Coyle et al.,

    2002). Jiang and Kang (2004) have suggested that gene therapy with

    MT could improve the recovery from liver fibrosis. The same

    authors have shown that mice which have developed a reversible

    liver fibrosis upon removal of CCl4, had a high level of hepatic MT,

    but mice which developed an irreversible fibrosis, had low

    expression of MT. Therefore, hepatic MT expression could reflect

    the severity of chronic liver damage, showing down-regulation in

    injured tissue (Carrera et al., 2003). The results of this study show

    that the MT I/II expression was progressively down-regulated from

    the CCl4 group to the CCl4 control group. The level of MTs in livers

    was related to the progression of CCl4-induced fibrosis and a

    complete down-regulation of MT I/II in the CCl4 control group

    indicates development of an irreversible fibrosis. On the other hand,

    the positive correlation of MT I/II expression with luteolin dosage

    and liver tissue repairment suggests its direct involvement in the

    regeneration of liver tissue. The liver sections from mice treated

    with luteolin 50 mg/kg have shown strong up-regulation of MT I/II,

    including nuclear expression, which was accompanied with a

    complete resolution of fibrotic scar tissue, suggesting the pro-nounced therapeutic effect of luteolin. Our results support the view

    that MT I/II could be the factors associated with a favorable clinical

    outcome in the response to therapy (Carrera et al., 2003).

    Regardless of these interesting and important findings, there are

    some limitations of the present study. The mechanistic insights into

    the luteolin-induced recovery of irreversible liver fibrosis need to be

    further approached. Although the important parameters of fibrosis

    and liver regeneration were determined, other factors that are

    possibly involved in the liver fibrinolysis need to be explored.

    In conclusion, the luteolin treatment has induced a complete

    reversion of established liver fibrosis by decreasing hepatic fibrogenic

    potential, and stimulated MT I/II expression in the liver in a dose-

    dependent manner, suggesting the strong enhancement of hepatic

    regenerative capability. Importantly, the results obtained from thisstudy suggest the therapeutic application of luteolin in patients with

    hepatic fibrosis, although further studies, in a more chronic model of

    liver fibrosis, are required.

    Conflict of interest statement

    The authors declare no conflict of interest.

    Acknowledgments

    This research was supported by grants from the Ministry of

    Science, Education and Sport, Republic of Croatia (project no. 062-

    0000000-3554). The authors thank Ing. Hrvoje Krian, Ljubica rnac,

    and Jadranka Ekinja for their excellent technical support and Dipl.

    Ing. Sunica Ostoji for her assistance with SDS-PAGE.

    320 R. Domitrovi et al. / Toxicology and Applied Pharmacology 241 (2009) 311321

  • 8/2/2019 Liver Fibrosis in Mice Induced by Carbon Tetra Chloride

    11/11

    References

    Andrews, G.K., 2000. Regulation of metallothionein gene expression by oxidative stressand metal ions. Biochem. Pharmacol. 59, 95104.

    Apostolova, M.D., Cherian, M.G., 2000. Nuclear localization of metallothionein duringcell proliferation and differentiation. Cell. Mol. Biol. 46, 347356.

    Arthur, M.J., 1994. Degradation of matrix proteins in liver fibrosis. Pathol. Res. Pract.190, 825833.

    Benyon, R.C., Iredale, J.P., 2000. Is liver fibrosis reversible? Gut 46, 443446.Bergman, A., Loxley, R., 1963. Two improved and simplified method for the

    determination of hydroxyproline. Anal. Chem. 35, 19611964.

    Blomhoff, R., Wake, K., 1991. Perisinusoidal stellate cells of the liver: important roles inretinol metabolism and fibrosis. FASEB J. 5, 271277.Blouin, A., Bolender, R.P., Weibel, E.R., 1977. Distribution of organelles and membranes

    between hepatocytes and nonhepatocytes in the rat liver parenchyma. A stereo-logical study. Cell. Biol. 72, 441455.

    Bradford, M.M., 1976. A rapid and sensitive method for the quantitation of microgramquantities of protein utilizing the principle of protein-dye binding. Anal. Biochem.72, 248254.

    Campbell, J.S., Hughes, S.D., Gilbertson, D.G., Palmer, T.E., Holdren, M.S., Haran, A.C.,Odell,M.M.,Bauer, R.L., Ren, H.P., Haugen, H.S., Yeh, M.M., Fausto, N.,2005.Platelet-derived growth factor C induces liver fibrosis, steatosis, and hepatocellularcarcinoma. Proc. Natl. Acad. Sci. U. S. A. 102, 33893394.

    Carrera, G., Paternain, J.L., Carrere, N., Folch, J., Courtade-Sadi, M., Orfila, C., Vinel, J.P.,Alric,L., Pipy, B.,2003. Hepatic metallothionein in patients with chronic hepatitisC:relationship with severity of liver disease and response to treatment. Am. J.Gastroenterol. 98, 11421149.

    Cassiman,D., Libbrecht, L.,Desmet, V.,Denef, C.,Roskams,T., 2002. Hepatic stellate cell/myofibroblast subpopulations in fibrotic human and rat livers. J. Hepatol. 36,200209.

    Cherian, M.G., Kang, Y.J., 2006. Metallothionein and liver cell regeneration. Exp. Biol.Med. (Maywood) 231, 138144.

    Chubatsu, L.S., Meneghini, R., 1993. Metallothionein protects DNA from oxidativedamage. Biochem. J. 291, 193198.

    Coleta, M., Campos, M.G., Cotrim, M.D., Lima, T.C., Cunha, A.P., 2008. Assessment ofluteolin (3,4,5,7-tetrahydroxyflavone) neuropharmacological activity. Behav.Brain Res. 189, 7582.

    Coyle, P., Philcox, J.C., Carey, L.C., Rofe, A.M., 2002. Metallothionein: the multipurposeprotein. Cell. Mol. Life Sci. 59, 627647.

    Denk, H., Stumptner, C., Zatloukal, K., 2000. Mallory body revisited. J. Hepatol. 32,689700.

    Domitrovi, R., Jakovac, H., Grebi, D., Milin, ., Radoevi-Stai, B., 2008a. Dose- andtime-dependent effects of luteolin on liver metallothioneins and metals incarbon tetrachloride-induced hepatotoxicity in mice. Biol. Trace Elem. Res. 126,176185.

    Domitrovi, R., Tota, M., Milin, ., 2008b. Differential effect of high dietary iron onalpha-tocopherol and retinol levels in the liver and serum of mice fed olive oil- andcorn oil-enriched diets. Nutr. Res. 28, 263269.

    Domitrovi, R., Jakovac, H., Milin, ., Radoevi-Stai, B., 2009. Dose- and time-dependent effects of luteolin on carbon tetrachloride-induced hepatotoxicity inmice. Exp. Toxicol. Pathol. doi:10.1016/j.etp.2008.12.005.

    Dooley, S., Hamzavi, J., Ciuclan, L., Godoy, P., Ilkavets, I., Ehnert, S., Ueberham, E.,Gebhardt, R., Kanzler, S., Geier, A., 2008. Hepatocyte-specific smad7 expressionattenuates TGF--mediated fibrogenesis and protects against liver damage.Gastroenterology 135, 642659.

    Friedman, S.L., 1999. Cytokines and fibrogenesis. Semin. Liver Dis. 9, 129140.Friedman, S.L., 2000. Molecular regulation of hepatic fibrosis, an integrated cellular

    response to tissue injury. J. Biol. Chem. 275, 22472250.Guyot, C., Lepreux, S., Combe, C., Doudnikoff, E., Bioulac-Sage, P., Balabaud, C.,

    Desmoulire, A., 2006. Hepatic fibrosis and cirrhosis: the (myo)fibroblastic cellsubpopulations involved. Int. J. Biochem. Cell. Biol. 38, 135151.

    Hagberg, H., Dammann, O., Mallard, C., Leviton, A., 2004. Preconditioning and thedeveloping brain. Semin. Perinatol. 28, 389395.

    Han, Y.P., 2006. Matrix metalloproteinases, the pros and cons, in liver fibrosis.J. Gastroenterol. Hepatol. 21, S8891.

    Havsteen, B.H., 2002. The biochemistry and medical significance of the flavonoids.Pharmacol. Ther. 96, 67202.

    Hosotani, K., Kitagawa, M., 2003. Improved simultaneous determination method ofbeta-carotene and retinol with saponification in human serum and rat liver.

    J. Chromatogr. B 791, 305313.Iredale, J.P., Benyon, R.C., Pickering, J., McCullen, M., Northrop, M., Pawley, S., Hovell, C.,

    Arthur, M.J., 1998. Mechanisms of spontaneous resolution of rat liver fibrosis.Hepatic stellate cell apoptosis and reduced hepatic expression of metalloproteinaseinhibitors. J. Clin. Invest. 102, 538549.

    Iredale, J.P., 2001. Hepatic stellate cell behavior during resolution of liver injury. Semin.Liver Dis. 21, 427436.

    Issa, R., Zhou, X., Constandinou, C.M., Fallowfield, J., Millward-Sadler, H., Gaca, M.D.,Sands,E., Suliman, I.,Trim,N., Knorr, A.,Arthur, M.J., Benyon, R.C., Iredale, J.P., 2004.Spontaneous recovery from micronodular cirrhosis: evidence for incompleteresolution associated with matrix cross-linking. Gastroenterology 126, 17951808.

    Jiang, Y., Kang, Y.J., 2004. Metallothionein gene therapy for chemical-induced liverfibrosis in mice. Mol. Ther. 10, 11301139.

    Ju, W., Wang, X., Shi, H., Chen, W., Belinsky, S.A., Lin, Y., 2007. A critical role of luteolin-induced reactive oxygen species in blockage of tumor necrosis factor-activatednuclear factor-kappaB pathway and sensitization of apoptosis in lung cancer cells.Mol. Pharmacol. 71, 13811388.

    Kinnman, N., Hultcrantz, R., Barbu, V., Rey, C., Wendum, D., Poupon, R., Housset, C.,2000. PDGF-mediated chemoattraction of hepatic stellate cells by bile ductsegments in cholestatic liver injury. Lab. Invest. 80, 697707.

    Knittel, T., Mehde, M., Kobold, D., Saile, B., Dinter, C., Ramadori, G., 1999. Expressionpatterns of matrix metalloproteinases and their inhibitors in parenchymal andnon-parenchymal cells of rat liver: regulation by TNF-alpha and TGF-beta1.

    J. Hepatol. 30, 4860.Knittel, T., Mehde, M., Grundmann, A., Saile, B., Scharf, J.G., Ramadori, G., 2000.

    Expression of matrix metalloproteinases and their inhibitors during hepatic tissuerepair in the rat. Histochem. Cell Biol. 113, 443453.

    Kopito, R.R., 2000. Aggresomes, inclusion bodies and protein aggregation. Trends Cell.

    Biol. 10, 524

    530.Kossakowska, A.E., Edwards, D.R., Lee, S.S., Urbanski, L.S., Stabbler, A.L., Zhang, C.L.,Phillips, B.W., Zhang, Y., Urbanski, S.J., 1998. Altered balance between matrixmetalloproteinases and their inhibitors in experimental biliary fibrosis. Am. J.Pathol. 153, 18951902.

    Kuo, W.H., Yang, S.F., Chu, S.C., Lu, S.O., Chou, F.P., Hsieh, Y.S., 2003. Differentialinductions of matrix metalloproteinase-2 and -9 in host tissues during the growthof ascitic sarcoma 180 cells in mice. Cancer Lett. 189, 103112.

    Marra, F., Romanelli, R.G., Giannini, C., Failli, P., Pastacaldi, S., Arrighi, M.C., Pinzani, M.,Laffi, G., Montalto, P., Gentilini, P., 1999. Monocyte chemotactic protein-1 as achemoattractant for human hepatic stellate cells. Hepatology 29, 140148.

    Mermelstein, C.S., Guma, F.C., Mello, T.G., Fortuna, V.A., Guaragna, R.M., Costa, M.L.,Borojevic, R., 2001. Induction of the lipocyte phenotype in murine hepatic stellatecells: reorganisation of the actin cytoskeleton. Cell. Tissue Res. 306, 7583.

    Miles, A.T., Hawksworth, G.M., Beattie, J.H., Rodilla, V., 2000. Induction, regulation,degradation and biological significance of mammalian metallothionein. Crit. Rev.Biochem. Mol. Biol. 35, 3570.

    Murayama, H., Ikemoto, M., Fukuda, Y., Tsunekawa, S., Nagata, A., 2007. Advantage ofserum type-I arginase and ornithine carbamoyltransferase in the evaluation of

    acute and chronic liver damage induced by thioacetamide in rats. Clin. Chim. Acta.375, 6368.

    Natarajan, S.K., Thomas, S., Ramachandran, A., Pulimood, A.B., Balasubramanian, K.A.,2005. Retinoid metabolism during development of liver cirrhosis. Arch. Biochem.Biophys. 443, 93100.

    Neubauer, K., Knittel, T., Aurisch, S., Fellmer, P., Ramadori, G.,1996. Glialfibrillary acidicproteina cell type specific marker for Ito cells in vivo and in vitro. J. Hepatol. 24,719730.

    Parola, M., Robino, G., 2001. Oxidative stress-related molecules and liver fibrosis.J. Hepatol. 35, 297306.

    Perez-Garcia, F., Adzet, T., Canigueral, S., 2000. Activity of artichoke leaf extract onreactive oxygen species in human leucocytes. Free Radical Res. 33, 661665.

    Perez-Tamayo, R., Montfort, I., Gonzalez, E., 1987. Collagenolytic activity in experi-mental cirrhosis of the liver. Exp. Mol. Pathol. 47, 300308.

    Poli, G., 2000. Pathogenesis of liver fibrosis: role of oxidative stress. Mol. Aspects Med.21, 4998.

    Radice, S., Marabini, L., Gervasoni, M., Ferraris, M., Chiesara, E., 1998. Adaptation tooxidative stress: effects of vinclozolin and iprodione on the HepG2 cell line.Toxicology 129, 183191.

    Salguero Palacios, R., Roderfeld, M., Hemmann, S., Rath, T., Atanasova, S., Tschuschner,A., Gressner, O.A., Weiskirchen, R., Graf, J., Roeb, E., 2008. Activation of hepaticstellate cells is associated with cytokine expression in thioacetamide-inducedhepatic fibrosis in mice. Lab. Invest. 88, 11921203.

    Seifert, W.F.,Bosma,A.,Brouwer,A., Hendriks,H.F.,Roholl,P.J., vanLeeuwen,R.E.,van Thiel-de Ruiter, G.C., Seifert-Bock, I., Knook, D.L., 1994. Vitamin A deficiency potentiatescarbon tetrachloride-induced liver fibrosis in rats. Hepatology 19, 193201.

    Shimoi, K., Okada, H., Furugori, M., Goda, T., Takase, S., Suzuki, M., Hara, Y., Yamamoto,H., Kinae, N., 1998. Intestinal absorption of luteolin and luteolin 7-O--glucoside inrats and humans. FEBS Lett. 438, 220224.

    Stumptner, C., Fuchsbichler, A., Heid, H., Zatloukal, K., Denk, H., 2002. Mallory body Adisease-associated type of sequestosome. Hepatology 35, 10531062.

    Tsukamoto, H., 1999. Cytokine regulation of hepatic stellate cells in liver fibrosis.Alcohol., Clin. Exp. Res. 23, 911916.

    Veda, H., Yamazaki, C., Yamazaki, M., 2002. Luteolin as an anti-inflammatory and anti-allergic constituent of Perilla frutescens. Biol. Pharm. Bull. 25, 11971202.

    Williams, T., Burk, R.F., 1990. Carbon tetrachloride hepatotoxicity: an example of freeradical-mediated injury. Semin. Liver Dis. 10, 279284.

    Wostowski, T., 1993. Involvement of metallothionein and copper in cell proliferation.BioMetals 6, 7176.Woessner, J.F.,1991. Matrix metalloproteinases and theirinhibitorsin connective tissue

    remodelling. FASEB J. 5, 21452154.Woodman, O.L., Chan, E.C.H., 2004. Vascular and anti-oxidant actions offlavonols and

    flavones. Clin. Exp. Pharmacol. Physiol. 31, 786790.Yang, C.,Zeisberg, M.,Mosterman, B.,Sudhakar, A.,Yerramalla,U., Holthaus,K., Xu,L., Eng,

    F.,Afdhal,N., Kalluri,R., 2003. Liverfibrosis:insightsinto migration of hepatic stellatecells in response to extracellular matrix and growth factors. Gastroenterology 124,147159.

    Yokoi,Y., Namihisa,T., Kuroda, H.,Komatsu,I., Miyazaki,A., Watanabe,S., Usui, K.,1984.Immunocytochemical detection of desmin in fat-storingcells (Itocells). Hepatology4, 709714.

    Zeisberg, M., Yang, C., Martino, M., Duncan, M.B., Rieder, F., Tanjore, H., Kalluri, R., 2007.Fibroblasts derive from hepatocytes in liver fibrosis via epithelial to mesenchymaltransition. J. Biol. Chem. 282, 2333723347.

    Ziyan, L., Yongmei, Z., Nan, Z., Ning, T., Baolin, L., 2007. Evaluation of the anti-inflammatory activity of luteolin in experimental animal models. Planta Med 73,221226.

    321R. Domitrovi et al. / Toxicology and Applied Pharmacology 241 (2009) 311321