toxicologic pathology - snubi.orgjuhan/pubfiles/methylenedianilin(toxicolpathol).pdfsociety of...

15
http://tpx.sagepub.com Toxicologic Pathology DOI: 10.1177/0192623308320272 2008; 36; 660 originally published online Jul 22, 2008; Toxicol Pathol Hyung-Lae Kim, Ju Han Kim, Heekyoung Chung, Gu Kong, Kyung-Sun Kang and Byung-IL Yoon Sun-Bom Kwon, Joon-Suk Park, Jung-Yeon Yi, Jae-Wong Hwang, Mingoo Kim, Mi-Ock Lee, Byung-Hoon Lee, 4,4'-methylene Dianiline, in Mouse Liver in an Acute Phase Time- and Dose-based Gene Expression Profiles Produced by a Bile-ductdamaging Chemical, http://tpx.sagepub.com/cgi/content/abstract/36/5/660 The online version of this article can be found at: Published by: http://www.sagepublications.com On behalf of: Society of Toxicologic Pathology can be found at: Toxicologic Pathology Additional services and information for http://tpx.sagepub.com/cgi/alerts Email Alerts: http://tpx.sagepub.com/subscriptions Subscriptions: http://www.sagepub.com/journalsReprints.nav Reprints: http://www.sagepub.com/journalsPermissions.nav Permissions: at Seoul National University on January 5, 2009 http://tpx.sagepub.com Downloaded from

Upload: nguyenkien

Post on 09-Jul-2018

224 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Toxicologic Pathology - snubi.orgjuhan/pubFiles/methyleneDianilin(ToxicolPathol).pdfSociety of Toxicologic Pathology Additional services and information for Toxicologic Pathology can

http://tpx.sagepub.com

Toxicologic Pathology

DOI: 10.1177/0192623308320272 2008; 36; 660 originally published online Jul 22, 2008; Toxicol Pathol

Hyung-Lae Kim, Ju Han Kim, Heekyoung Chung, Gu Kong, Kyung-Sun Kang and Byung-IL Yoon Sun-Bom Kwon, Joon-Suk Park, Jung-Yeon Yi, Jae-Wong Hwang, Mingoo Kim, Mi-Ock Lee, Byung-Hoon Lee,

4,4'-methylene Dianiline, in Mouse Liver in an Acute PhaseTime- and Dose-based Gene Expression Profiles Produced by a Bile-duct�damaging Chemical,

http://tpx.sagepub.com/cgi/content/abstract/36/5/660 The online version of this article can be found at:

Published by:

http://www.sagepublications.com

On behalf of:

Society of Toxicologic Pathology

can be found at:Toxicologic Pathology Additional services and information for

http://tpx.sagepub.com/cgi/alerts Email Alerts:

http://tpx.sagepub.com/subscriptions Subscriptions:

http://www.sagepub.com/journalsReprints.navReprints:

http://www.sagepub.com/journalsPermissions.navPermissions:

at Seoul National University on January 5, 2009 http://tpx.sagepub.comDownloaded from

Page 2: Toxicologic Pathology - snubi.orgjuhan/pubFiles/methyleneDianilin(ToxicolPathol).pdfSociety of Toxicologic Pathology Additional services and information for Toxicologic Pathology can

660

Time- and Dose-based Gene Expression Profiles Producedby a Bile-duct–damaging Chemical, 4,4’-methylene Dianiline,

in Mouse Liver in an Acute Phase

SUN-BOM KWON,1 JOON-SUK PARK,2 JUNG-YEON YI,1 JAE-WONG HWANG,2 MINGOO KIM,3 MI-OCK LEE,4,8 BYUNG-HOON LEE,5,8 HYUNG-LAE KIM,6,8 JU HAN KIM,3,8 HEEKYOUNG CHUNG,7,8 GU KONG,7,8 KYUNG-SUN KANG,2,8 AND BYUNG-IL YOON1,8

1 School of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University,Chuncheon 200-701, Republic of Korea

2 Department of Veterinary Public Health, College of Veterinary Medicine, Seoul National University,Seoul 151-742, Republic of Korea

3 Seoul National University Biomedical Informatics (SNUBI) and Human Genome Research Institute,College of Medicine, Seoul National University, Seoul 110-799, Republic of Korea

4 College of Pharmacy and Bio-MAX Institute, Seoul National University, Seoul 151-742, Republic of Korea5 College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University,

Seoul 151-742, Republic of Korea6 Department of Biochemistry, College of Medicine, Ewha Women’s University, Seoul 158-710, Republic of Korea

7 College of Medicine, Hanyang University, Seoul 133-891, Republic of Korea8 Toxicogenomics Research Consortium (TGRC), Hanyang University, Seoul 133-891, Republic of Korea

ABSTRACT

A toxicogenomics study was performed in the mouse liver after treatment of a bile-duct–damaging chemical, 4,4’-methylene dianiline (MDA), acrossmultiple doses and sampling times in an acute phase using the AB Expression Array System. Imprinting control region (ICR) mice were given a singleoral administration of a low (10 mg/kg b.w.) or high (100 mg/kg b.w.) dose of MDA. Mice were sacrificed six, twenty-four, and seventy-two hours aftertreatment for serum chemistry, histopathology, and mRNA preparation from liver samples. Treatment with MDA increased liver-toxicity–relatedenzymes in blood and induced bile-duct cell injury, followed by regeneration. To explore potential biomarker gene profiles, the altered genes werecategorized into four expression patterns depending on dose and time. Numerous functionally defined and unclassified genes in each category wereup- or down-regulated throughout the period from cellular injury to the recovery phase, verified by RT-PCR. Many genes associated with liver toxi-city and diseases belonged to one of these categories. The chemokine-mediated Th1 pathway was implicated in the inflammatory process. The genesassociated with oxidative stress, apoptosis, and cell-cycle regulation were also dynamically responsive to MDA treatment. The Wnt/β-cateninsignaling pathway was likely responsible for the reconstitution process of the MDA-injured liver.

Keywords: acute liver toxicity; 4,4’-methylene dianiline; mouse; toxicogenomics.

produced by chemicals can therefore provide practical infor-mation to not only predict their potential toxicity in specific tis-sues, but also to understand the molecular mechanismsinvolved (Bartosiewicz et al. 2001; Walker et al. 2006).

The liver is vulnerable to a variety of metabolic, toxic,microbial, circulating, and neoplastic insults. Therefore,microenvironmental changes in liver tissues will reflect the bio-chemical characteristics and potential toxicity of certain chem-icals and compounds. Understanding such changes is importantfor risk assessment in humans and animals. Toxicogenomicsstudies have focused on identifying gene expression changes inthe liver following treatment with hepatotoxicants, whichinduce hepatocytic cell injury (Bartosiewicz et al. 2001; Huanget al. 2004). However, very few genomics studies have exam-ined altered gene expression profiles induced by bile-duct-cell–damaging chemicals.

INTRODUCTION

The intra- and intercellular microenvironmental changesinduced by a variety of toxicants are characterized by theirunique gene expression profiles, which makes it possible toidentify the expression signatures of altered genes followingtreatment with different chemicals (Hamadeh et al. 2002;Huang et al. 2004). The unique gene expression profiles

Sun-Bum Kwon and Joon-Suk Park contributed equally to this work.Address correspondence to: Byung-IL Yoon, DVM, PhD, School of Veterinary

Medicine, Kangwon National University, 192-1 Hyoja2-dong, Chuncheon,Kangwon 200-701, Republic of Korea; e-mail: [email protected].

Abbreviations: AB, Applied Biosystems; ALT, alanine aminotransferase;AST, aspartate aminotransferase; COA, correspondence analysis; DEA, differ-ential expression analysis; MDA, 4,4’-methylene dianiline; NO, nitric oxide;ROS, reactive oxygen species.

Toxicologic Pathology, 36: 660-673, 2008Copyright © 2008 by Society of Toxicologic PathologyISSN: 0192-6233 print / 1533-1601 onlineDOI: 10.1177/0192623308320272

at Seoul National University on January 5, 2009 http://tpx.sagepub.comDownloaded from

Page 3: Toxicologic Pathology - snubi.orgjuhan/pubFiles/methyleneDianilin(ToxicolPathol).pdfSociety of Toxicologic Pathology Additional services and information for Toxicologic Pathology can

Vol. 36, No. 5, 2008 TOXICOGENOMICS BY ACUTE TREATMENT OF 4,4’-METHYLENE DIANILINE IN MOUSE LIVER 661

4,4’-methylene dianiline (MDA) is used in the synthesis ofisocyanates and polyurethane polymers and in the productionof dyes, polycondensation products, copolymers, curatives forepoxy resins, and other compounds (Kanz et al. 1992). Bile-duct cells are the main target cells of this chemical, whichinduces cholangitis with cellular necrosis and inflammation,resulting in “epping jaundice” in humans (Kanz et al. 1992;Kopelman et al. 1966).

In the present study, we performed a toxicogenomics studyto identify the distinct gene expression profiles altered byMDA in an acute phase. The aim of this study is to extend ourunderstanding of the possible mechanisms by which MDAinduces bile-duct damage, followed by cellular recovery, at thelevel of global genes and to identify the potential target geneprofiles for risk assessment and classification of the chemicalsor compounds that cause liver toxicity, especially those likeMDA that cause toxicity toward bile-duct cells.

MATERIALS AND METHODS

Materials

MDA (CAS #101-77-9), chloroform, RNase-free 75%ethanol, RNase-free distilled water (DEPC water), and iso-propyl alcohol were all purchased from Aldrich Sigma (St.Louis, MO, USA). RNAlater RNA stabilization solution wasobtained from Ambion Inc. (Austin, TX, USA). TRIzol andRNeasy Mini kits for RNA isolation were purchased fromInvitrogen (Carlsbad, CA, USA) and Qiazen (Valencia, CA,USA), respectively.

Animals and Chemical Treatment

Specific pathogen-free (SPF) slc:ICR mice were purchasedfrom Charles River Laboratories of Japan. The mice were housedat five per polycarbonate cage in an approved animal facility ofthe College of Veterinary Medicine of Seoul National University,Korea. Mice were maintained on a twelve-hour light-dark cycle.A basic pellet diet and water were provided ad libitum through-out the study. Temperature and humidity inside the raising roomwere maintained automatically at 23°C ± 2°C and 50% ± 20%,respectively. The mice were acclimatized for one week prior tothe beginning of the study. All animals used in this study weretreated humanely according to the guidelines of the InstitutionalAnimal Care and Use Committee (IACUC) at Seoul NationalUniversity for compliance with the National Research Council’sGuide for the Care and Use of Laboratory Animals (NationalAcademic Press 1996). Animals were randomly divided intothree groups: a vehicle control and two experimental groups. Twodifferent doses of MDA were determined based on histopatho-logical data from preliminary experiments; 100 mg/kg b.w. and10 mg/kg b.w. In the preliminary study, 100 mg/kg b.w. (selectedas the high-dose group) was the lowest dose showing typical inju-rious morphological changes of the bile ducts. Because it isimportant that with low doses, cellular injury is difficult to detectby light microscopic examination, 10 mg/kg b.w. was alsoselected as the lower dose group where no morphological

changes were observed; it was also the geometric ratio of thehigh dose.

After one week of acclimatization, the mice were subjected toexperimental treatments. Food was withdrawn for four hoursbefore treatment (from 8:00 AM to 1:00 PM). Thereafter the miceof each dose group, including the vehicle group, were given asingle oral administration of the selected dose of chemicalsand/or vehicles. Based on the data of the preliminary study,which showed that twenty-four hours post-MDA single treatmentwas the peak time point of MDA-induced liver injury, three sam-pling time points were determined to observe the altered geneexpression profiles at the early (six hours post-treatment) and therecovery (seventy-two hours post-treatment) phases, as well asthe injury peak time point (twenty-four hours post-treatment).Animals were provided a normal diet (Purina Inc., Korea) andtap water ad libitum throughout the experimental period.

Histopathology and Serum Chemistry

The mice were sacrificed after collecting blood from the abdom-inal artery under ether anesthesia. Liver tissues were removed andthen fixed in 10% neutral buffered formalin. After routine tissue pro-cessing, the tissues were embedded in low-melting-point paraffin.Tissue sections of 3 μm were stained with hematoxylin and eosin(H & E) for histological examination.

Serum was prepared from the collected blood and analyzedusing chemical analyses for aspartate aminotransferase (AST),alanine aminotransferase (ALT), alkaline phosphatase, albumin,total bilirubin, blood urea nitrogen, creatinine, and triglyceride.

RNA Preparation and Microarray Analysis Usingthe Applied Biosystems Expression Array System

Three animals out of five in each group were selected asrepresentative animals based on biochemical and histopatho-logical review. Total RNA was extracted from the left lobe ofthe collected liver tissues from the three selected mice of eachgroup using TRIzol and RNeasy Mini kits. The purity and qual-ity of the isolated RNA samples were analyzed using an AgilentBioanalyzer 2110 (Agilent Technologies, Santa Clara, CA,USA) to confirm that the 28S/18S ratio was between 1.8 and 2.0and the 260/280 nm ratio was between 2.0 and 2.2, respectively.The quality of the isolated RNA samples was further deter-mined by electrophoresis on denaturing formaldehyde–agarosegels. Applied Biosystems (AB, Foster City, CA, USA) MouseGenome Survey Arrays were used to analyze gene expressionprofiles following the single administration of MDA and/orvehicle. The AB mouse genome survey array contains 33,315probes, representing 32,381 curated genes targeting 44,498transcripts. The usefulness and effectiveness of the ABmicroarray were validated in previous studies (Walker et al.2006). Digoxigenin-UTP–labeled cRNA was generated andlinearly amplified from 1 μg of total RNA from each sampleusing an AB Chemiluminescent RT-IVT Labeling Kit (version1.5). Array hybridization chemiluminescence detection andimage acquisition and analysis were performed using the AB

at Seoul National University on January 5, 2009 http://tpx.sagepub.comDownloaded from

Page 4: Toxicologic Pathology - snubi.orgjuhan/pubFiles/methyleneDianilin(ToxicolPathol).pdfSociety of Toxicologic Pathology Additional services and information for Toxicologic Pathology can

662 KWON ET AL. TOXICOLOGIC PATHOLOGY

Chemiluminescence Detection Kit and the AB 1700Chemiluminescent Microarray Analyzer. Images were auto-gridded, then spotted and spatially normalized. Signals werequantified and corrected for background noise, and the finalimages and feature data were processed using AB 1700Chemiluminescent Microarray Analyzer software (version1.1). Data and images were collected through an automatedprocess for each microarray using the 1700 analyzer. Generalinformation about the strategy for microarray data qualityassurance in the ABI microarray system is available athttp://www.appliedbiosystems.com/.

Statistical Analysis of Microarray Data

Microarray data were analyzed using the software Avadis 3.3prophetic (Strand Genomics Pvt Ltd.). The local background wassubtracted from the raw expression values for all spots. The ratioswere then log-transformed (base 2) and normalized so that themedian log-transformed ratio equaled zero. Gene expressionratios were centered on the median across all samples. Theexpression ratio of each gene was determined by dividing the nor-malized expression value of a gene in a chemical treatment groupby the mean expression value in the vehicle control group foreach sampling time. Genes indicating a change of more than 1.5-fold were usually included in the data analysis. We used thesupervised analysis method for differential expression analysis(DEA). Differential expression analysis was done between thevehicle control and chemical treatment for each sampling time.DEA was performed using Avadis software. Permutation-basedmodified t tests were used to provide further confidence in theseresults. Differential gene expression was analyzed using a two-sample Welch Benjamin Hochberg t statistic. Thus, differentialgene expression associated with each group was tested using thesignificance analysis of microarrays.

Gene expression values were manipulated and visualizedusing the R package (free software under the terms of the FreeSoftware Foundation’s General Public License). For the analy-sis of data correlation, correspondence analysis was also per-formed using the AB1700 package in R. Hierarchical clusteranalysis partitioned the data into discrete hierarchical groupsbased on data trends. The resulting gene lists were limited togenes with changes in proportions of 1.5 or more and p < .05.

Categorization of Microarray Gene Expression Profiles

Because our goal was to define gene expression profiles topredict liver toxicity, especially associated with bile duct cellinjury, we categorized altered genes based on the expression pat-terns represented at the selected sampling times in the differentdose groups (Figure 1) and further classified them by function.

Semiquantitative RT-PCR

To verify whether the genes identified from the microarrayresults were altered by MDA treatment, RT-PCR analyses were

performed for twelve genes, as described previously (Park et al.2004) using specific primers (Table 1). Equal amounts of totalmRNA from the three samples selected from each group werepooled. First-strand cDNA was generated from 2 μg of totalRNA by RT with 1 μL of 50 μM oligo (dT)20 primer and 1 μLof SuperScript III Reverse Transcriptase (Life TechnologiesInc., Gaithersburg, MD, USA) in a 20-μL reaction mixture, fol-lowing the manufacturer’s instructions. From the 20 μL pro-duced, PCR was performed using AccuPower PCR PreMix(Bioneer, Daejeon, Korea) and using a Mycycler (Bio-Rad,Hercules, CA, USA). The PCR products were analyzed by elec-trophoresis in 1.5% agarose gels containing 0.1 mg/mL ethid-ium bromide. Images of RT-PCR ethidium-bromide–stainedagarose gels were acquired with ultraviolet illumination on aGel Doc EQ System (Bio-Rad, Hercules, CA, USA).

RESULTS

Histopathology and Serum Chemistry

In the low-dose group, no notable histopathological changeswere observed for any of the sampling times (Figure 2). In thehigh-dose group, MDA-induced cellular toxicity was limitedmainly to the bile duct cells (Figure 2). Six hours after treat-ment, bile duct epithelial cells showed hydropic degenerationand, sometimes, necrotic changes. Necrosis of bile duct cells,resulting in exfoliation of the epithelial cells from the basementmembrane, was evident twenty-four hours after the high-dosetreatment of MDA. Seventy-two hours after the high-dosetreatment of MDA, bile duct epithelial cells were regenerative,which was characterized by large basophilic epithelial cells oftall cuboidal shape and relatively frequent mitotic figures. Aperiductular desmoplastic reaction was also prominent, withthe infiltration of a number of mononuclear cells. Individualnecrosis and multiple necrotic foci of hepatocytes wereobserved randomly in the liver.

Serum chemical analyses indicated significant increases inAST, ALT, and total bilirubin twenty-four hours after treatmentwith the high dose of MDA, followed by a return toward con-trol values at seventy-two hours (Table 2). With the low dose ofMDA, no significant changes in the parameters examined wereobserved for any sampling time.

Gene Expression Changes

After quantile-normalization of the raw data, statisticalmicroarray analyses were conducted. First, we verified a goodcorrelation (> 95%) of the gene expression profile betweenindividuals in the same group for each sampling time. Wholemicroarray data are found at http://www.snubi.org/publication/TGRC_MDA. The low and high doses of MDA produced dif-ferential gene expression profiles for each selected samplingtime. In the hierarchical clustering, the gene expression profileswere likely to be affected by the sampling time, rather than thedose of MDA, until twenty-four hours after treatment.However, at seventy-two hours, the gene expression profileswere distinctly comparable between the low- and high-dose

at Seoul National University on January 5, 2009 http://tpx.sagepub.comDownloaded from

Page 5: Toxicologic Pathology - snubi.orgjuhan/pubFiles/methyleneDianilin(ToxicolPathol).pdfSociety of Toxicologic Pathology Additional services and information for Toxicologic Pathology can

Vol. 36, No. 5, 2008 TOXICOGENOMICS BY ACUTE TREATMENT OF 4,4’-METHYLENE DIANILINE IN MOUSE LIVER 663

groups (Figure 3). Two-way ANOVA of the dose- and time-based microarray data identified 4,257 altered genes in the low-dose group and 4,534 genes in the high-dose group, all withstatistically significant changes (p < .05). These microarraydata were further filtered into a list of genes with changes morethan 1.5 times the control level. The numbers of altered genes

(p < .05, with a change of more than two times) for each sam-pling time in the low- and high-dose groups are shown in Figure4. Categorization of the altered genes based on the PANTHERbiological process indicated that a variety of genes were impli-cated in the biological processes of the liver after MDA treatment,associated with cell cycle and proliferation; cell structure and

FIGURE 1.—Strategy for the classification of genes with altered expression.

TABLE 1.—Primer sequences used to validate the microarray data by semiquantitative RT-PCR.

Spot ID Gene Sense primer Antisense primer

472781 Dio1 GGAACCATAGGCATTGGAAA TGCCATGGAAACACCTATCA578302 Prss22 GACAGTGGGGACACAGGAAG CCCCGGAGGTAGAAGACAT578137 Havcr1 CCGTGTCTCTGCTCACTACAG CCAACTTTTAATTCCTATCCCTCT701888 Cox10 AGCAGGAAGCGTATTTTGGT GTTTGGGAAGCAGTTTGGAT752346 Sult3a1 CATAATTCACAGACAAAATGATTACC GAAACCAATGCTAGTAGCTCCAT847906 Raf1 TCAGATGATGGCAAGCTCAC GCACATTGACCACAGTCCTT509971 Bgn CTGGAGAACAGTGGCTTTGA TGTTGTGGTCCAGGTGAAGT524988 Egr1 TCAAGCAGCAGTCCTAGGTATTA AACCCAACCAAACAAACCAT355859 Nmyc1 TAATGAGAGGTGGCTCTTGC TACAAAACATGCCCCTCAAA759375 Cxcl1 TTTGTATGTATTAGGGTGAGGACAT CACGTGCGTGTTGACCATA743516 Mybbp1a ATGACGCTGATGAGAAGCAG TTCACTTTCTTCCCCATCCT648614 Galk2 GGGTCCACACCCAAGTTTTA TTTTCACAGGTTCCACAGCTA

at Seoul National University on January 5, 2009 http://tpx.sagepub.comDownloaded from

Page 6: Toxicologic Pathology - snubi.orgjuhan/pubFiles/methyleneDianilin(ToxicolPathol).pdfSociety of Toxicologic Pathology Additional services and information for Toxicologic Pathology can

664 KWON ET AL. TOXICOLOGIC PATHOLOGY

motility; developmental processes; immunity and defense; intra-cellular protein traffic; lipid, fatty acid, and steroid metabolism;protein metabolism and modification; signal transduction;transport; and so on (Figure 5). The global gene expression pro-files were classified further based on the comparative geneexpression patterns between the low- and high-dose groups(Figure 1). Semiquantitative RT-PCR was performed using spe-cific primers (Table 1) to verify the gene expression profilesdetected using the oligonucleotide microarray. The semiquantita-tive RT-PCR and microarray results were quite compatible foreach sampling time: ten of twelve genes (83.3%) at six hours,nine of twelve genes (75.0%) at twenty-four hours, and ten oftwelve genes (83.3%) at seventy-two hours showed the samepatterns of change (Figure 6).

Class I: Genes That Showed Similar Expression Patterns at theLow and High Dose: This class contains genes that were up- ordown-regulated in both the low- and high-dose groups withsimilar patterns of expression (Supplementary Table 1: http://www.snubi.org/publication/TGRC_MDA). Various genes asso-ciated with amino acid metabolism; apoptosis; carbohydratemetabolism; the cell cycle; cell structure and motility; develop-mental processes; electron transport; immunity and defense;lipid, fatty acid, and steroid metabolism; nucleoside, nucleotide,and nucleic acid metabolism; oncogenesis; signal transduction;and other functions were placed in this category. In particular,the genes that were altered concomitantly with both the lowand high doses were found most frequently at the peak injurytime twenty-four hours after treatment with MDA.

FIGURE 2.—Histopathology of mice sacrificed at six (2A, 2B, 2C), twenty-four (2D, 2E, 2F), and seventy-two hours (2G, 2H, 2I) after vehiclecontrol (2A, 2D, 2G), low (2B, 2E, 2H), and high (2C, 2F, 2I) methylene dianiline (MDA) treatment. Throughout the study, no specific patho-logical changes were observed in the bile ducts at the low dose (10 mg/kg), but typical bile duct damage (arrows in 2C and 2F), followed by bileduct cell regeneration (arrow in 2I) and fibrosis with inflammation, was noted at the high dose (100 mg/kg) of MDA. Bar = 10 μm, except 20 μmin 2H. H & E stain.

at Seoul National University on January 5, 2009 http://tpx.sagepub.comDownloaded from

Page 7: Toxicologic Pathology - snubi.orgjuhan/pubFiles/methyleneDianilin(ToxicolPathol).pdfSociety of Toxicologic Pathology Additional services and information for Toxicologic Pathology can

Vol. 36, No. 5, 2008 TOXICOGENOMICS BY ACUTE TREATMENT OF 4,4’-METHYLENE DIANILINE IN MOUSE LIVER 665

The genes associated with the induction of apoptosis, forexample, Chordc1, Tnfrsf6, and Casp3, were up-regulated irre-spective of dose. Several notable genes were associated withcarbohydrate metabolism: Gbe1, Galk2, and Galnt11. Gbe1 isup-regulated in the hypoxic environmental conditions of themouse liver (Zhao et al. 2004). In association with the cellcycle, cyclin D1 was concomitantly down-regulated at the latephase, whereas other cell cycle-related genes, including Cks2,Nudc, Mobk1b, and Rback, were up-regulated at twenty-fourhours after MDA treatment. The alteration of cortactin, whichis involved in cytoskeletal reorganization (Lua et al. 2005), wasnotable because it was highly up-regulated by more than tentimes at twenty-four hours after MDA treatment for both doselevels. Genes associated with developmental processes andelectron transport tended to be down-regulated by MDA treat-ment, especially at the time of peak injury at twenty-four hours.In contrast, many genes associated with intracellular proteintraffic and lipid, fatty acid, and steroid metabolism were up-regulated. In particular, Copg and Sgpl1 (apoptosis-associatedgenes under stress) were prominently elevated and Gpx6 wasaffected at the peracute phase.

Class II: Genes That Showed Similar Expression Patterns at AnySampling Time(s) at Both Dose Levels, but Showed Dose-dependent Expression at Any Other Sampling Time(s): This class contains genes that were dose-independently up- or down-regulated at any sampling time, but also showed dose-dependent expression at any other sampling time (SupplementaryTable 2: http://www.snubi.org/publication/TGRC_MDA). Thegenes in this class generally showed up- or down-regulation attwenty-four hours for both the low and high doses and reversealteration at seventy-two hours for the high dose. For instance,arginase, which is involved in nitric oxide (NO) and reactiveoxygen species (ROS) metabolism, was down-regulated attwenty-four hours for both doses, but was up-regulated at sev-enty-two hours in the high-dose treatment. Many genes postu-lated to be associated with liver diseases, including biliary

diseases, belonged to this class (Table 3). Anapc5, which playsan important role in the regulation of unscheduled hepatocytecell proliferation (Wirth et al. 2004), was prominently overex-pressed at twenty-four hours for both doses (12.3- and 9.3-foldchanges in low and high doses, respectively) but was dramati-cally down-regulated at the regenerating phase for the highdose. Raf1 showed an expression pattern similar to that ofAnapc5. These genes appeared to play important roles in recon-stitution processes after MDA-induced liver injury. Emid2 andLepr were noteworthy as genes with rapid responses to MDAtreatment.

Class III: Genes That Showed Reverse Expression PatternsBetween the Low and High Dose: This class contains genes thatwere down-regulated for the low dose and up-regulated for thehigh dose, and vice versa (Supplementary Table 3: http://www.snubi.org/publication/TGRC_MDA). These genes mayreflect differential responses of the liver depending on intensityand type of cellular injury. Catalase was included in this cate-gory; it was highly overexpressed at twenty-four hours for thelow dose, but it significantly down-regulated at the active regen-erating phase for the high dose, suggesting the importance ofoxidative stress in association with toxic mechanisms and thepreventive function of catalase at a low dose of MDA. Ehpb1,Map3k4, Mipep, Hsd3b7, Slc25a25, and Slc25a15 showed thesame expression patterns as catalase. In contrast, Galnt7, B4galt4,and Mt1 had expression patterns opposite to that of catalase. Theywere down-regulated for the low dose, but they were highly over-expressed at the regenerating phase for the high dose.

Class IV: Genes That Were Separately Affected in an On/OffPattern Dose-dependently: The genes in this class may repre-sent differential and separate genetic responses in proportion tothe dose of MDA (Supplementary Table 4: http://www.snubi.org/publication/TGRC_MDA. In association with apopto-sis, at the low dose, Bcl-2 and related genes appeared to functionin maintaining cellular homeostasis, whereas at the high dose,

TABLE 2.—Serum chemistry of the mice from each group at the selected sampling time points (mean value ± SD).

Six hours Twenty-four hours Seventy-two hours

Group VC Low High VC Low High VC Low High

AST 85.0 ± 14.3 91.8 ± 19.7 69.8 ± 9.0 85.2 ± 23.5 89.6 ± 21.2 756.4 ± 174.7* 74.6 ± 31.9 75.4 ± 33.2 166.2 ± 131.7*ALT 28.4 ± 6.11 33.8 ± 5.26 29.0 ± 4.08 28.3 ± 11.8 33.4 ± 9.56 331.2 ± 354.2* 25.3 ± 6.26 29.4 ± 10.6 151.8 ± 226.5*ALP 339.8 ± 106.1 402.8 ± 88.8 331.8 ± 24.7 437.3 ± 90.2 325.6 ± 101.7 276.4 ± 24.5 307.0 ± 77.3 316.4 ± 74.6 388.0 ± 37.7T-BIL 1.06 ± 0.28 1.22 ± 0.32 1.37 ± 1.00 0.93 ± 0.37 0.98 ± 1.30 2.06 ± 1.82 1.33 ± 0.54 1.20 ± 0.38 1.90 ± 1.19BUN 22.4 ± 3.01 20.8 ± 1.94 21.6 ± 1.70 85.2 ± 23.5 18.4 ± 2.60 23.8 ± 3.00 23.6 ± 3.44 24.02 ± 2.36 25.02 ± 2.32CREA 0.46 ± 0.04 0.47 ± 0.03 0.44 ± 0.01 0.50 ± 0.02 0.50 ± 0.03 0.43 ± 0.04 0.52 ± 0.05 0.49 ± 0.04 0.47 ± 0.09TG 95.6 ± 10.04 122.6 ± 32.4 135.3 ± 37.9 118.0 ± 27.9 140.4 ± 17.3 117.8 ± 54.2 120.8 ± 16.6 144.8 ± 27.7 150.6 ± 36.4ALB 2.98 ± 0.18 3.04 ± 0.15 2.93 ± 0.98 2.45 ± 1.05 3.04 ± 0.09 2.86 ± 0.21 3.00 ± 0.07 3.04 ± 0.11 3.00 ± 0.12

Note: The mice of each group were sacrificed at six, twenty-four, and seventy-two hours after a single administration of vehicle control (VC), 10 mg/kg b.w. (Low) and 100 mg/kg b.w.(High) of 4,4’-methylene dianiline.

Abbreviations: AST, aspartate aminotransferase; ALT, alanine aminotransferase; ALP, alkaline phosphatase; ALB, albumin;T-BIL, total bilirubin; BUN, blood urea nitrogen; CREAT,creatinine; TG, triglyceride.

* Significant difference from the respective vehicle control group at p < .05.

at Seoul National University on January 5, 2009 http://tpx.sagepub.comDownloaded from

Page 8: Toxicologic Pathology - snubi.orgjuhan/pubFiles/methyleneDianilin(ToxicolPathol).pdfSociety of Toxicologic Pathology Additional services and information for Toxicologic Pathology can

666 KWON ET AL. TOXICOLOGIC PATHOLOGY

the alteration of the apoptosis-inhibiting genes Angptl4 andBirc5, and the apoptosis-induction–associated genes Lgals4,Foxo1, Foxo3, Tnfrsf23, Ing1l, and Pdcd4 was noted, with thedown-regulation of caspase 3 at the regenerating phase. Theexpression of cell-cycle–related genes was very differentdepending on the dose of MDA. At the low dose, the cell cyclecheckpoint genes cyclin G1 and G2 were up-regulated, but atthe high dose, cyclin B2, Cdc20, Cks1, Mad2l1, and Prc1 wereup-regulated at the regenerating phase. However, cyclin D3

was rather depressed by the high dose of MDA at the regener-ating phase. At the peak injury time of the high-dose group,marked up-regulation of Pak4 (p21-activated kinase 4) wasnoted. In association with immunity and defense, thechemokine ligand series genes were usually up-regulated atboth the low and high doses. In particular, marked up-regulationof Cxcl1 by 11.1 times at the peracute phase was notable in thelow-dose group. Histocompatibility-2–related genes were acti-vated in the low-dose group but down-regulated in the high-dose group. Lymphocyte-related genes were usually activatedat the late phase in the high-dose group, possibly reflectinglymphocyte-mediated chronic inflammation, which was alsonoted in histopathological findings (Figure 2). In addition,Sod1, an endogenous antioxidant, was significantly activated inthe high-dose group. Numerous genes associated with nucleo-side, nucleotide, and nucleic acid metabolism belonged in thisclass. These genes were usually found at the early and peakinjury times in the low-dose group and at the late phase in thehigh-dose group. In particular, in the low-dose group, Egr1(early growth response 1) was dramatically up-regulated by44.5 times at the peracute phase, possibly associated with liverregeneration and cholestasis (Table 3) (Liao et al. 2004; Kimet al. 2006). Many ribosomal proteins responded differently toMDA, depending on the dose. They were usually down-reg-ulated at the early phase at the low dose, whereas they weredown-regulated at the peak injury time followed by up-regulationin the regenerating phase at the high dose.

SUMMARY AND DISCUSSION

MDA is a selective bile duct toxicant (Kanz et al. 1992), asalso indicated by our histopathological observations in an asso-ciation with serum chemistry. However, the MDA-inducedtoxic effect is achieved not only by targeting biliary cells, butalso by breaking the functional relationships with other livercell components. In the genomic circumstances, host cellresponses against chemical-inducing effects are even moredynamic and active, but very much conserved. Thus, ourmicroarray data represented complicated and understandableexpression changes in a variety of genes maintaining cellularmicroenvironmental homeostasis across MDA-induced toxiceffects and adaptation in the liver, including the genes alteredin response to bile duct cell injury by MDA.

Exploration of Potential Biomarker Gene Profiles toDiscern the Toxic Endpoints of Bile Duct Cells

The gene expression profiles produced by oral treatmentwith MDA differed greatly depending on the dose and sam-pling time after treatment. This finding reflects the highlydynamic genetics that maintain cellular homeostasis despiteacute cellular injury. Based on the comparative gene expressionpatterns between the low and high doses, MDA-affected geneswere classified into four categories, Classes I to IV. Classes Iand II contained genes affected by MDA independent of thedose, whereas Classes III and IV contained entirely dose-dependent genes.

FIGURE 3.—Hierarchical cluster analysis of differentially altered genesin methylene dianiline (MDA)-treated mouse liver in the acute phase,which was made from the averaged signal of three microarray datafrom each time and dose group. In the matrix, red and green indicategenes that were up- and down-regulated by MDA treatment, respec-tively, for each sampling time.

at Seoul National University on January 5, 2009 http://tpx.sagepub.comDownloaded from

Page 9: Toxicologic Pathology - snubi.orgjuhan/pubFiles/methyleneDianilin(ToxicolPathol).pdfSociety of Toxicologic Pathology Additional services and information for Toxicologic Pathology can

Vol. 36, No. 5, 2008 TOXICOGENOMICS BY ACUTE TREATMENT OF 4,4’-METHYLENE DIANILINE IN MOUSE LIVER 667

The genes belonging to Classes I and II could be affected bychemicals similar to MDA independent of dose. In fact, severalgenes that are postulated to be associated with liver toxicity,including bile duct damage, were frequently found in theseclasses (Table 3). Class I and II genes may also represent themicroenvironmental conditions associated with the toxic mech-anism of MDA. Cortactin (Cttn) and catenin beta (Catnb) arefunctionally involved in cell mortility (Chuma et al. 2004; Luaand Low 2005) and Wnt signaling (Janssens et al. 2006),respectively, and will contribute to the remodeling of theMDA-injured liver together with cell-cycle– and apoptosis–regulating genes such as cyclin D, Rback, caspase3, sphingosinephosphate lyase 1, and so on (Supplementary Table 3:http://www.snubi.org/publication/TGRC_MDA). An iron home-ostatic gene, Alas1, is also activated by MDA treatment, whichis associated with heme synthesis regulated by bile acids in theliver (Peyer et al. 2007). In contrast, the genes belonging toClasses III and IV could be potential biomarker genes in pre-dicting the level of exposure to MDA-like chemicals becausetheir expression was dose dependent. As shown in Class III, manygenes that were altered at the time of peak injury (i.e., twenty-fourhours) in the low-dose group had reversed expression patterns

during the recovery phase (i.e., seventy-two hours) in the high-dose group. Metallothionein-1 and catalase have been shown tobe altered by some liver toxicants such as furan (Bartosiewiczet al. 2001; Huang et al. 2004). In contrast, genes belonging toClass IV were affected in an on/off dose-dependent expressionpattern. Among the genes affected only by the low dose of MDA,some genes such as Sdh1 (2.5-times increase), Gck (4.1-timesincrease), and Hyal (2.1-times decrease followed by 3.0-timesincrease) have already been postulated as biochemical markersassociated with liver function and toxicity, including biliary dis-eases (Table 3). In addition, Caveolin 2 (Cav2), which is highlyexpressed in the proliferating bile ductules in primary biliary cir-rhosis (Yokomori et al. 2005), was significantly up-regulated at thetime of peak injury at the high dose of MDA. In association withliver fibrosis, overexpression of smad 1 was noteworthy at the latestage of severely MDA-injured liver (Table 3) (Fan et al. 2006).Glypican 3, a cell-adhesion–related gene, is a useful marker forhepatocellular carcinoma (Libbrecht et al. 2006), but MDA,a bile-duct-damaging chemical, induced glypican 1 at the lowdose and glypicans 4 and 5 at the high dose.

Early-response genes have been an important topic in toxi-cogenomics studies. In the microarray, Egr1, Nmyc1, Cxcl1,

FIGURE 4.—Venn diagram showing the number of genes altered more than two-fold during methylene dianiline-induced liver injury and recovery.

at Seoul National University on January 5, 2009 http://tpx.sagepub.comDownloaded from

Page 10: Toxicologic Pathology - snubi.orgjuhan/pubFiles/methyleneDianilin(ToxicolPathol).pdfSociety of Toxicologic Pathology Additional services and information for Toxicologic Pathology can

668 KWON ET AL. TOXICOLOGIC PATHOLOGY

Clrf, Hmgcs1, P2rx2, Gng2, and Dnajb4 were consideredpotential early biomarkers associated with biliary cell damagein liver exposed to a low dose of MDA because they weredramatically up-regulated at the early phase (six hours) inthe low-dose group. In particular, Egr1 was up-regulated byapproximately forty-five times in the early phase after the lowdose of MDA. This gene changes rapidly at the mRNA level inthe livers of mice with cholestasis (Kim et al. 2006). Galk2(galactokinase 2), which is regulated by Egr1 (Yang et al.2004), was subsequently up-regulated by low and high doses ofMDA. More importantly, Gpx6 (+2.5 times, +2.9 times),Emid2 (+2.8, +2.8), Nr0b2 (+4.1, +3.2), Slc23a3 (+3.2, +2.3),Dio1 (+5.5, +5.0), Ard1 (–2.4, –3.0), Prss22 (–5.3, –5.8), And(–2.2, –2.5), Wbscr16 (–2.6, –3.3), Havcr1 (–4.8, –4.5), Cox10(–2.2, –2.9), and other genes showed marked alterations at boththe low and high doses in the early phase after MDA treatment.The altered expression of these genes was further verified usingRT-PCR in cases (Figure 6).

Comprehensive Action Mechanisms ofAltered Genes Following MDA Treatment

Wnt Signaling Pathway: The Wnt genes are involved in celldifferentiation, proliferation, maturation, and cell fate determi-nation by regulating gene expression (Janssens et al. 2006).Thus, they play important roles in homeostatic mechanisms inadult tissues. Based on the microarray, the Wnt/β-catenin sig-naling pathway is most likely to play important roles in the

reconstitution of MDA-injured liver (Figure 7). The first genesactivated by MDA treatment were Nmyc1, Tle1 (transducin-like enhancer of split 1), Gng2 (guanine nucleotide bindingprotein gamma 2), and Pcdha8 (protocadherin alpha 8).Following the activation of these was the activation of β-catenin and its downstream genes, such as the protein phos-phatase family (Ppp2ca, Ppp3ca, and calcineurin B), cadherin,calcineurin, SWI/SNF, Tle3 (transducin-like enhancer of split3), Btaf1 (BTAF1 RNA polymerase II), Acvr1 (activin A recep-tor), and Pcdhga12 (protocadherin gamma subfamily A 12).Myconcogenes were activated only by high doses of MDA. Incontrast, Wnt2, cyclin D1, and Prkcz (protein kincase C zeta)were down-regulated in the late recovery phase after MDAtreatment, presumably representing a feedback response to reg-ulate cell proliferation.

Immunity and Inflammation: In the high-dose group, periductalinflammation characterized by lymphocyte infiltration washistopathologically evident in the acute late phase. This findingwas supported by the up-regulation of numerous immunity-associated genes. Lymphocyte-related genes such as Lsp1, Ly6a,and Ly96, and chemokine-related genes such as Ccl2, Ccl9,Cxcl9, and Xcl1 were up-regulated, implying that the Th1 path-way was involved in MDA-induced inflammation. The activationof these types of proinflammatory cytokines also occurs in micesuffering from biliary atresia (Leonhardt et al. 2006). Further, reg-ulators of cytokine signaling such as Jak1 and Map3k4 were alsoactivated by MDA treatment.

FIGURE 5.—Gene ontology of the biological processes for the methylene dianiline (MDA)-modulated genes. Analysis based on the biologicalprocesses of PANTHER indicated that several functional genes are implicated in the biological cellular processes in the liver after MDA treat-ment. Numbers on the Y axis represent the number of altered genes.

at Seoul National University on January 5, 2009 http://tpx.sagepub.comDownloaded from

Page 11: Toxicologic Pathology - snubi.orgjuhan/pubFiles/methyleneDianilin(ToxicolPathol).pdfSociety of Toxicologic Pathology Additional services and information for Toxicologic Pathology can

Vol. 36, No. 5, 2008 TOXICOGENOMICS BY ACUTE TREATMENT OF 4,4’-METHYLENE DIANILINE IN MOUSE LIVER 669

FIGURE 6.—Semiquantitative RT-PCR and microarray results of selected clones. Each probe is represented by its gene symbol (left of the panel),and the corresponding spot ID is in parentheses. Numbers at the top of the gel are from microarray data of the indicated spot ID, which are givenas the median time change (log2; n = 3) relative to the time-matched, vehicle-treated control. RT-PCR was performed using randomly selected RNAsamples from each experimental group. The experiments were repeated at least three times, and representative figures are shown. V, vehicle-treatedcontrol; L, low dose of methylene dianiline (MDA); H, high dose of MDA.

at Seoul National University on January 5, 2009 http://tpx.sagepub.comDownloaded from

Page 12: Toxicologic Pathology - snubi.orgjuhan/pubFiles/methyleneDianilin(ToxicolPathol).pdfSociety of Toxicologic Pathology Additional services and information for Toxicologic Pathology can

670 KWON ET AL. TOXICOLOGIC PATHOLOGY

Cell Cycle and Proliferation: Cell proliferation after acuteinjury by MDA was achieved via pathways associated withinsulin-like growth factor and epidermal growth factor. Thep53-gene–mediated pathways seemed to play an important rolein cell-cycle regulation during the reconstitution of liverwith bile duct cell damage. In the recovery phase, which wascharacterized histopathologically by the regenerative prolifera-tion of bile duct epithelial cells and periductal fibroplasia,Cyclin B2, Cdc20, and Mad2 were highly up-regulated,whereas Cdkn2b (a cyclin-dependent kinase inhibitor), Anapc5(a regulator of unscheduled cell proliferation), and Gadd45gwere down-regulated, indicating cell cycle progression in agree-ment with histopathological observations (Figure 2). Cyclin B2,as a regulatory subunit of the maturation-promoting factor, isconsidered to be associated with the formation of the bipolarspindle at metaphase during cell meiosis (Kotani et al. 2001).However, the G1 progression genes cyclin D1 and D3 weredown-regulated during the recovery phase. In particular, cyclinD1 was inhibited by MDA at both the low and high doses. Cell-cycle regulation also appeared to be achieved by RB and itsrelated genes, as supported by the high expression of Rbak (RB-associated KRAB repressor) and Rbbp9. The altered expressionof Cul4a (cullin 4A), which is involved in the MDM2-mediatedproteolysis of p53 (Nag et al. 2004), also suggests a regulatory

role in cell proliferation in MDA-injured liver. In contrast, CyclinG1 and G2, which are cell-cycle arrest genes mediated by p53(Harris and Levine 2005), were activated twenty-four hours aftera low dose of MDA, which might be associated with conservedcell-cycle regulation by feedback loops in cases of reversibleinjury without serious cellular damage.

Apoptosis: Programmed cell death is a process that precedestissue reconstruction and causes irreversibly damaged cells tobe removed. A single dose of MDA induced the activationof the apoptosis-inducing genes Chordc1 and Tnfrsf6 and theup-regulation of caspase 3 at twenty-four hours in both thelow- and high-dose groups. In the low-dose group, Bcl-2–related genes seemed to play key roles in the regulation ofcell death. In the high-dose group, apoptosis-inducing and -inhibiting genes were counterbalanced (Lgals4, Foxo1, andIng1l vs. Angptl4 and Birc5).

Oxidative Stress: Several endogenous antioxidant genes andredox-regulating genes showed altered expression after MDAtreatment. Gpx6 (glutathione peroxidase 6), catalase, andTxndc1 (thioredoxin domain-containing 1) were significantlyactivated in the low-dose group. The increased expression ofthese antioxidant enzymes suggests that treatment with MDA

TABLE 3.—Genes postulated to be associated with liver diseases and their expression patterns in the liverafter a single oral administration of 4,4′-methylene dianiline.

Low(10 mg/kg) High(100 mg/kg)

Gene name Class Associated liver diseases Reference 6 h 24 h 72 h 6 h 24 h 72 h

Galk2 I Glycogen storage disease Brivet et al. 1983 NC 4.6 NC NC 4.3 NCAlas1 I Iron homeostasis in liver Peyer et al. 2007 NC 3.6 2.7 NC 3.5 NCCortactin I Cell motility and migration Chuma et al. 2004 NC 15.6 NC NC NC 12.4Arg 1 II Obstructive jaundice Beck et al. 2005 NC –1.8 NC NC –1.7 2.1

CCl4-induced hepatic injury Mendez et al. 2006Idh1 II Toxic hepatitis Andreeshcheva et al. 2006 NC 3.6 NC NC 2.7 –4.5Spnb2 II Primary biliary cirrhosis Mishra et al. 2004 NC 3.7 NC NC 3.0 –3.3Nrf1 II Nonalcoholic steatohepatitis and hepatic neoplasia Zhenrong et al. 2005 NC 3.1 NC NC 2.8 –2.8Scd1 II Cholestasis Flowers et al. 2006 –2.3 2.6 2.4 NC NC –2.0Adk II Neonatal hepatic steatosis Boison et al. 2002 NC 2.6 NC NC 2.3 –2.7Ahcyl1 II Alcoholic liver injury Kharbanda et al. 2005 NC 7.9 NC NC 6.3 –3.4Pcsk9 II Hypercholesterolemia Maxwell et al. 2005 NC 5.8 NC NC 7.7 –7.2Ide II Hepatic cirrhosis Ono et al. 1984 NC 4.3 NC NC 3.2 –3.4Bgn* II Progressive liver fibrosis Hogemann et al. 1997 NC 4.5 NC NC 5.0 –4.9Ephb1 III Cholangiocyte pathophysiology Ueno et al. 2007 NC 2.7 NC NC NC –2.8Cat III Chemical-induced liver toxicity Huang et al. 2004 NC 2.8 NC NC NC –5.3Mt1 III Chemical-induced liver toxicity Bartosiewicz et al. 2001 NC -3.3 NC NC NC 5.5Egr-1* IV Liver regeneration Liao et al. 2004 44.5 NC NC NC NC NCHyal1 IV DMN-induced liver toxicity, early stage George and Stern 2004 –2.1 3.0 NC NC NC NC

Biliary cirrhosis Nyberg et al. 1988Sdh1 IV Liver toxicity Ozer et al. 2008 NC 2.5 NC NC NC NCGck IV Glucose homeostasis Matschinsky 2005 NC 4.1 NC NC NC NCSmad1 IV Liver fibrosis Fan et al. 2006 NC NC NC NC NC 3.6Smad7 IV Liver fibrosis Seyhan et al. 2006 NC NC –2.7 NC NC NC

The numbers indicate changes in gene expression relative to the gene expression in the corresponding vehicle control group. Abbreviation: NC, no change. * indicate the gene verified by RT-PCR in Figure 6

at Seoul National University on January 5, 2009 http://tpx.sagepub.comDownloaded from

Page 13: Toxicologic Pathology - snubi.orgjuhan/pubFiles/methyleneDianilin(ToxicolPathol).pdfSociety of Toxicologic Pathology Additional services and information for Toxicologic Pathology can

Vol. 36, No. 5, 2008 TOXICOGENOMICS BY ACUTE TREATMENT OF 4,4’-METHYLENE DIANILINE IN MOUSE LIVER 671

may induce oxidative stress in the liver, particularly at a lowdose. In contrast, at the high dose, catalase was markedlydown-regulated, which agreed with toxicogenomic results forother biliary toxic chemicals such as methapyrilene and furanin the liver (Huang et al. 2004). A decrease in catalase activityoccurs in liver exposed to oxidative-stress–inducible chemicalsand toxins such as 2-nitropropane and heptapeptide toxinmicrocystins (Borges et al. 2006; Jayaraj et al. 2006). Theimportance of catalase in chemical-induced liver toxicity isstrongly supported by its preventive effect against carbon tetra-chloride (CCl4) toxicity (Ma et al. 2006). At the high dose ofMDA, the redox-regulating system seemed to be disturbed,because Gpx1, Gpx5, and thioredoxin reductase 1 (Txbrd1), aswell as catalase, were significantly suppressed. Thus, oxidativestress production and redox system disruption may be otherimportant ways in which MDA induces liver toxicity.

In summary, a single oral administration of MDA produceddifferential gene expression profiles in mouse liver tissues thatwere dependent on or independent of sampling time and dose.With regard to potential biomarker genes, we divided thealtered genes into four categories, Classes I to IV. Numerousfunctionally defined and unclassified genes in each categorywere differentially up- or down-regulated throughout theperiod from cellular injury to the recovery phase at the exam-ined doses, providing potential biomarker gene profiles. Thesegene expression profiles were also comprehensive with regardto the mechanistic responses maintaining cellular homeostasisor recovery from MDA-induced liver toxicity. Thus, our resultsare valuable not only for understanding the mechanisms bywhich MDA-like chemicals induce liver toxicity, in particularbile duct cell injury, but also for risk assessment and classifica-tion of the chemicals using biotoxchips.

FIGURE 7.—Wnt/β-catenin signaling pathway. The red- and blue-colored genes represent the up- and down-regulated genes by acute treatment ofMDA, respectively. Based on the microarray, the Wnt/β-catenin signaling pathway is likely to play important roles in the reconstitution of MDA-injured liver.

at Seoul National University on January 5, 2009 http://tpx.sagepub.comDownloaded from

Page 14: Toxicologic Pathology - snubi.orgjuhan/pubFiles/methyleneDianilin(ToxicolPathol).pdfSociety of Toxicologic Pathology Additional services and information for Toxicologic Pathology can

672 KWON ET AL. TOXICOLOGIC PATHOLOGY

ACKNOWLEDGMENT

This work was supported by a grant from the Korea Foodand Drug Administration (KFDA-05122-TGP-584).

REFERENCES

Andreeshcheva, E. M., Popova, T. N., Artiukhov, V. G., Rakhmanova, T. I., andMatasova, L. V. (2006). The intensity of free radical oxidation and cat-alytic properties of the rat liver NADP-isocitrate dehydrogenase in thenorm and in toxic hepatitis. Biomed Khim 52, 153–60.

Bartosiewicz, M. J., Jenkins, D., Penn, S., Emery, J., and Buckpitt, A. (2001).Unique gene expression patterns in liver and kidney associated withexposure to chemical toxicants. J Pharmacol Exp Ther 297, 895–905.

Beck, B., Ciszek, M., Polaniak, R., Beyga, Z., Krol, W., Drozdz, M., and Shani,J. (2005). The activity of ornitine transcarbamoylase and arginase duringmechanical jaundice in the rat model. J Surg Res 126, 19–26.

Boison, D., Scheurer, L., Zumsteg, V., Rülicke, T., Litynski, P., Fowler, B.,Brandner, S., and Mohler, H. (2002). Neonatal hepatic steatosis by dis-ruption of the adenosine kinase gene. Proc Natl Acad Sci USA 99,6985–90.

Borges, L. P., Nogueira, C. W., Panatieri, R. B., Batista, J., Rocha, T., and Zeni,G. (2006). Acute liver damage induced by 2-nitropropane in rats: effectof diphenyl diselenide on antioxidant defenses. Chem Biol Interact 160,99–107.

Brivet, M., Moatti, N., Corriat, A., Lemonnier, A., and Odievre, M. (1983).Defective galactose oxidation in a patient with glycogen storage diseaseand Fanconi syndrome. Pedatr Res 17, 157–61.

Chuma, M., Sakamoto, M., Yasuda, J., Fujii, G., Nakanishi, K., Tsuchiya, A.,Ohta, T., Asaka, M., and Hirohashi, S. (2004). Overexpression of cor-tactin is involved in motility and metastasis of hepatocellular carcinoma.J Hepatol 41, 629–36.

Fan, J., Shen, H., Sun, Y., Li, P., Burczynski, F., Namaka, M., and Gong, Y.(2006). Bone morphogenetic protein 4 mediates bile duct ligation-induced liver fibrosis through activation of Smad1 and ERK1/2 in rathepatic stellate cells. J Cell Physiol 207, 499–505.

Flowers, M. T., Groen, A. K., Oler, A. T., Keller, M. P., Choi, Y., Schueler, K.L., Richards, O. C., Lan, H., Miyazaki, M., Kuipers, F., Kendziorski, C.M., Ntambi, J. M., and Attie, A. D. (2006). Cholestasis andhypercholesterolemia in SCD1-deficient mice fed a low-fat, high-carbohydrate diet. J Lipid Res 47, 2668–80.

George, J., and Stern, R. (2004). Serum hyaluronan and hyaluronidase: veryearly markers of toxic liver injury. Clin Chim Acta 348, 189–97.

Hamadeh, H. K., Bushel, P. R., Jayadev, S., Martin, K., DiSorbo, O., Sieber, S.,Bennett, L., Tennant, R., Stoll, R., Barrett, J. C., Blanchard, K., Paules, R.S., and Afshari, C. A. (2002). Gene expression analysis reveals chemical-specific profiles, Toxicol Sci 67, 219–31.

Harris, S. L., and Levine, A. J. (2005). The p53 pathway: positive and negativefeedback loops. Oncogene 24, 2899–980.

Hogemann, B., Edel, G., Schwarz, K., Krech, R., and Kresse, H. (1997).Expression of biglycan decorin and proteoglycan-100/CSF-1 in normaland fibrotic human liver. Pathol Res Pract 193, 747–51.

Huang, Q., Jin, X., Gaillard, E. T., Knight, B. L., Pack, F. D., Stoltz, J. H.,Jayadev, S., and Blanchard, K. T. (2004). Gene expression profilingreveals multiple toxicity endpoints induced by hepatotoxicants. Mut Res549, 147–68.

Janssens, N., Janicot, M., and Perera, T. (2006). The Wnt-dependent signalingpathways as target in oncology drug discovery. Invest New Drugs 24,263–80.

Jayaraj, R., Anand, T., and Rao, P. V. L. (2006). Activity and gene expressionprofile of certain antioxidant enzymes to microcystin-LR induced oxida-tive stress in mice. Toxicology 220, 136–46.

Kanz, M. F., Kaphalia, L., Kaphalia, B. S., Romagnoli, E., and Ansari, G. A.(1992). Methylene dianiline: acute toxicity and effects on biliary func-tion. Toxicol Appl Pharmacol 117, 88–97.

Kharbanda, K. K., Rogers, D. D. II, Mailliard, M. E., Siford, G. L., Barak, A.J., Beckenhauer, H. C., Sorrell, M. F., and Tuma, D. J. (2005). Role of

elevated S-adenosylhomocysteine in rat hepatocyte apoptosis: protectionby betaine. Biochem Pharmacol 70, 1883–90.

Kim, N. D., Moon, J. O., Slitt, A. L., and Copple, B. L. (2006). Early growthresponse factor-1 is critical for cholestatic liver injury. Toxicol Sci 90,586–95.

Kotani, T., Yoshida, N., Mita, K., and Yamashita, M. (2001). Requirement ofcyclin B2, but not cyclin B1, for bipolar spindle formation in frog (Ranajaponica) oocytes. Mol Reprod Dev 59, 199–208.

Kopelman, H., Scheuer, P. J., and Williams, R. (1966). The liver lesion of theEpping jaundice. Quart J Med 35, 553–64.

Leonhardt, J., Stanulla, M., von Wasielewski, R., Skokowa, J., Kübler, J., Ure,B. M., and Petersen, C. (2006). Gene expression profile of the infectivemurine model for biliary atresia. Pediatr Surg Int 22, 84–89.

Liao,Y., Shikapwashya, O. N., Shteyer, E., Dieckgraefe, B. K., Hruz, P. W., andRudnick, D. A. (2004). Delayed hepatocellular mitotic progression andimpaired liver regeneration in early growth response-1-deficient mice. JBiol Chem 279, 43107–16.

Libbrecht, L., Severi, T., Cassiman, D., Borght, S. V., Pirenne, J., Nevens, F.,Verslype, C., van Pelt, J., and Roskams, T. (2006). Glypican-3 expressiondistinguishes small hepatocellular carcinomas from cirrhosis, dysplasticnodules, and focal nodular hyperplasia-like nodules. Am J Surg Pathol30, 1405–11.

Lua, B. L., and Low, B. C. (2005). Cortactin phosporylation as a switch for actincytoskeletal network and cell dynamics control. FEBS Lett 579, 577–85.

Ma, S.-F., Nishikawa, M., Katsumi, H., Yamashita, F., and Hashida, M. (2006).Liver targeting of catalase by cationization for prevention of acute liverfailure in mice. J Control Release 110, 273–82.

Matschinsky F. M. (2005). Glucokinase, glucose homeostasis, and diabetesmellitus. Curr Diab Rep 5, 171–76.

Maxwell, K. N., and Breslow, J. L. (2005). Proprotein convertase subtilisinkexin 9: the third locus implicated in autosomal dominant hypercholes-terolemia. Curr Opin Lipidol 16, 167–72.

Mendez, J. D., De Haro Hernandez, R., and Conejo, V. A. (2006). Spermineincreases arginase activity in the liver after carbon tetrachloride-inducedhepatic injury in Long-Evans rats. Biomed Pharmacother 60, 82–85.

Mishra, B., Tang,Y., Katuri, V., Fleury, T., Said, A. H., Rashid, A., Jogunoori, W.,and Mishra, L. (2004). Loss of cooperative function of transforminggrowth factor-beta signaling proteins, smad3 with embryonic liver fodrin,a beta-spectrin, in primary biliary cirrhosis. Liver Int 24, 637–45.

Nag, A., Bagchi, S., and Raychaudhuri, P. (2004). Cul4A physically associateswith MDM2 and participates in the proteolysis of p53. Cancer Res 64,8152–55.

Nyberg, A., Engstrom-Laurent, A., and Loof, L. (1988). Serum hyaluronate inprimary biliary cirrhosis – a biochemical marker for progressive liverdamage. Hepatology 8, 142–46.

Ono, J., Hutson, D. G.., Dombro, R. S., Zeppa, R., Katsuki, T. (1984). Insulindegradation in hepatic cirrhosis. Gastroenterol Jpn 19, 99–103.

Ozer, J., Ratner, M., Shaw, M., Bailey, W. and Schomaker, S. (2008). The currentstate of serum biomarkers of hepatotoxicity. Toxicology 245, 194–205.

Park, J. S., Noh, D. Y., Kim, S. H., Kong, G., Chang, C. C., Lee, Y. S., Trosko,J. E., and Kang, K. S. (2004). Gene expression analysis in SV40-immor-talized human breast luminal epithelial cells with stem cells characteris-tics using a cDNA microarray. Int J Oncol 24, 1545–58.

Peyer, A. K., Jung, D., Beer, M., Gnerre, C., Keogh, A., Stroka, D., Zavolan, M.,and Meyer, U. A. (2007). Regulation of human liver delta-aminolevulinicacid synthase by bile acids. Hepatology 46, 1960–70.

Ueno, Y., Fukushima, K., Nakagome, Y., Kido, O., and Shimosegawa, T.(2007). Bioinformatic approach for cholangiocyte pathophysiology.Hepathol Res 37, S444–S448.

Walker, S. J., Wang, Y., Grant, K. A., Chan, F., and Hellmann, G. M.(2006). Long versus short oligonucleotide microarrays for the studyof gene expression in nonhuman primates. J Neurosci Methods 152,179–89.

Wirth, K. G., Ricci, R., Giménez-Abián, J. F., Taghybeeglu, S., Kudo, N. R.,Jochum, W., Vasseur-Cognet, M., and Nasmyth, K. (2004). Loss of theanaphase-promoting complex in quiescent cells causes unscheduledhepatocyte proliferation. Genes Dev 18, 88–98.

at Seoul National University on January 5, 2009 http://tpx.sagepub.comDownloaded from

Page 15: Toxicologic Pathology - snubi.orgjuhan/pubFiles/methyleneDianilin(ToxicolPathol).pdfSociety of Toxicologic Pathology Additional services and information for Toxicologic Pathology can

Vol. 36, No. 5, 2008 TOXICOGENOMICS BY ACUTE TREATMENT OF 4,4’-METHYLENE DIANILINE IN MOUSE LIVER 673

Yang, F., Agulian, T., Sudati, J. E., Rhoads, D. B., and Levitsky, L. L. (2004).Developmental regulation of galactokinase in suckling mouse liver by theEgr-1 transcription factor. Pediatr Res 55, 822–29.

Yokomori, H., Oda, M., Wakabayashi, G.., Kitajima, M., Yoshimura, K.,Nomura, M., and Hibi, T. (2005). High expressions of caveolins on the pro-liferating bile ductules in primary biliary cirrhosis. World J Gastroenterol11, 3710–13.

Zhao, J., Chen, H., Davidson, T., Kluz, T., Zhang, Q., and Costa, M. (2004).Nickel-induced 1,4-α-glucan branching enzyme 1 up-regulation via thehypoxic signaling pathway. Toxicol Appl Pharmacol 196, 404–9.

Zhenrong, X., Chen, L., Leung, L., Benedict Yen, T. S., Lee, C., and Chan, J. Y.(2005). Liver-specific inactivation of the Nrf1 gene in adult mouse leads to nonalcoholic steatohepatitis and hepatic neoplasia. PNAS 102,4120–25.

at Seoul National University on January 5, 2009 http://tpx.sagepub.comDownloaded from