24.01.2013 Views

Molecular characteristics of Kidney Cancer - KOPS - Universität ...

Molecular characteristics of Kidney Cancer - KOPS - Universität ...

Molecular characteristics of Kidney Cancer - KOPS - Universität ...

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

MOLECULAR CHARACTERISTICS OF<br />

KIDNEY CANCER<br />

DISSERTATION<br />

Zur Erlangung des akademischen Grades des<br />

Doktors der Naturwissenschaften an der <strong>Universität</strong> Konstanz<br />

Fachbereich Biologie<br />

Vorgelegt von<br />

KERSTIN STEMMER<br />

Konstanz, im Oktober 2008<br />

Tag der mündlichen Prüfung:<br />

15. Dezember 2008<br />

Referenten:<br />

Pr<strong>of</strong>. Dr. Daniel R. Dietrich<br />

Pr<strong>of</strong>. Dr. Christ<strong>of</strong> R. Hauck<br />

PD Dr. Sonja von Aulock<br />

I


“Success consists <strong>of</strong> going from failure to failure without loss <strong>of</strong> enthusiasm”<br />

Winston Churchill (1874-1965)<br />

III


Table <strong>of</strong> Contents<br />

Table <strong>of</strong> Contents........................................................................................................................................ IV<br />

List <strong>of</strong> Figures.............................................................................................................................................. XI<br />

List <strong>of</strong> Tables............................................................................................................................................. XIII<br />

Abbreviations ............................................................................................................................................XIV<br />

Publications and Presentations ..................................................................................................................XV<br />

Zusammenfassung..................................................................................................................................XVIII<br />

Summary...................................................................................................................................................XXI<br />

Chapter 1: Introduction........................................................................................................................... 1<br />

1.1 <strong>Kidney</strong> <strong>Cancer</strong>........................................................................................................................... 1<br />

1.1.1 Risk Factors for <strong>Kidney</strong> <strong>Cancer</strong>............................................................................................ 1<br />

1.1.2 Genetic and Epigenetic Causes <strong>of</strong> <strong>Cancer</strong>........................................................................... 2<br />

1.1.3 Models <strong>of</strong> Carcinogenesis .................................................................................................... 3<br />

1.1.4 Renal Tumor Pathology........................................................................................................ 4<br />

1.2 Renal Carcinogens.................................................................................................................. 10<br />

1.2.1 Genotoxic Carcinogens ...................................................................................................... 10<br />

1.2.2 Non-Genotoxic Carcinogens .............................................................................................. 11<br />

1.3 Renal Carcinogens Relevant for this Study............................................................................. 13<br />

1.3.1 Aristolochic acid.................................................................................................................. 13<br />

1.3.2 Ochratoxin A....................................................................................................................... 15<br />

1.3.3 Cycasin / Methylazoymethanol........................................................................................... 16<br />

1.4 Methods to Identify Renal Carcinogens................................................................................... 18<br />

1.4.1 Standard carcinogenicity testing......................................................................................... 19<br />

1.4.2 The lifetime rodent bioassay............................................................................................... 19<br />

1.4.3 The Eker Rat Model <strong>of</strong> Renal Carcinogenesis.................................................................... 21<br />

1.5 Toxicogenomics ...................................................................................................................... 22<br />

1.5.1 Principles <strong>of</strong> the Microarray Technology............................................................................. 22<br />

1.5.2 Microarrays- a Novel Tool to Study Carcinogenesis........................................................... 24<br />

1.5.3 Microarrays- Current Limitations and Future Directions ..................................................... 27<br />

Chapter 2: Work Hypotheses & Experimental Design.......................................................................... 28<br />

IV


2.1 Part I........................................................................................................................................ 28<br />

2.1.1 Experimental setup............................................................................................................. 29<br />

2.2 Part II....................................................................................................................................... 30<br />

2.2.1 Experimental setup:............................................................................................................ 30<br />

2.3 Part III...................................................................................................................................... 31<br />

2.3.1 Experimental setup............................................................................................................. 31<br />

2.4 Schematic Overview on the Experimental Design................................................................... 32<br />

Chapter 3: Manuscript I........................................................................................................................ 33<br />

3.1 Abstract ................................................................................................................................... 33<br />

3.2 Introduction ............................................................................................................................. 34<br />

3.3 Material and Methods.............................................................................................................. 36<br />

3.3.1 Compounds ........................................................................................................................ 36<br />

3.3.2 Animals............................................................................................................................... 36<br />

3.3.3 Animal treatment and sample collection ............................................................................. 36<br />

3.3.4 Histopathology.................................................................................................................... 37<br />

3.3.5 Immunohistochemistry........................................................................................................ 37<br />

3.3.6 RNA isolation and expression pr<strong>of</strong>iling ............................................................................... 37<br />

3.3.7 Microarray data processing and statistical analysis............................................................ 38<br />

3.3.8 Functional analysis <strong>of</strong> microarray data ............................................................................... 38<br />

3.3.9 Statistical analysis .............................................................................................................. 39<br />

3.4 Results .................................................................................................................................... 39<br />

3.4.1 Cell proliferation data.......................................................................................................... 39<br />

3.4.2 Preneoplastic and neoplastic pathology ............................................................................. 39<br />

3.4.3 Non- neoplastic pathology .................................................................................................. 40<br />

3.4.4 Gene expression pr<strong>of</strong>iles.................................................................................................... 42<br />

3.4.5 Functional analysis <strong>of</strong> significantly deregulated genes....................................................... 43<br />

3.4.6 Genes deregulated by aristolochic acid.............................................................................. 43<br />

3.4.7 Genes deregulated by ochratoxin A ................................................................................... 48<br />

3.5 Discussion............................................................................................................................... 57<br />

3.6 Supplement ............................................................................................................................. 62<br />

V


3.6.1 Supplementary Information ................................................................................................ 62<br />

3.6.2 Supplementary Tables........................................................................................................ 63<br />

Chapter 4: Manuscript II....................................................................................................................... 73<br />

4.1 Abstract ................................................................................................................................... 73<br />

4.2 Introduction ............................................................................................................................. 74<br />

4.3 Material and Methods.............................................................................................................. 76<br />

4.3.1 Compound.......................................................................................................................... 76<br />

4.3.2 Animals............................................................................................................................... 76<br />

4.3.3 Short-term experiments ...................................................................................................... 77<br />

4.3.4 Subchronic and chronic experiments.................................................................................. 77<br />

4.3.5 Sample collection ............................................................................................................... 77<br />

4.3.6 Histopathology.................................................................................................................... 77<br />

4.3.7 Cell proliferation analysis.................................................................................................... 78<br />

4.3.8 RNA isolation microarray hybridization and gene expression pr<strong>of</strong>iling ............................... 78<br />

4.3.9 Quantification <strong>of</strong> O6MG ...................................................................................................... 79<br />

4.3.10 Statistical analysis ......................................................................................................... 79<br />

4.4 Results .................................................................................................................................... 80<br />

4.4.1 Gene expression pr<strong>of</strong>iling................................................................................................... 80<br />

4.4.2 Non-neoplastic renal pathology .......................................................................................... 83<br />

4.4.3 (Pre)-neoplastic renal pathology......................................................................................... 84<br />

4.4.4 Cell proliferation.................................................................................................................. 87<br />

4.4.5 Detection <strong>of</strong> O6MG in MAMAc treated Eker-rats................................................................ 88<br />

4.5 Discussion............................................................................................................................... 89<br />

4.6 Supplement ............................................................................................................................. 92<br />

4.6.1 Supplementary experimental procedures: Real time PCR ................................................. 92<br />

4.6.2 Supplementary Figures ...................................................................................................... 92<br />

4.6.3 Supplementary Tables........................................................................................................ 93<br />

Chapter 5: Manuscript III...................................................................................................................... 95<br />

5.1 Abstract ................................................................................................................................... 95<br />

5.2 Introduction ............................................................................................................................. 96<br />

5.3 Material and Methods.............................................................................................................. 98<br />

VI


5.3.1 Animal treatment and tissue preparation ............................................................................ 98<br />

5.3.2 Sectioning and staining ...................................................................................................... 98<br />

5.3.3 Laser-assisted microdissection........................................................................................... 99<br />

5.3.4 RNA isolation and quality control........................................................................................ 99<br />

5.3.5 RNA amplification ............................................................................................................. 100<br />

5.3.6 Affymetrix chip hybridization and expression pr<strong>of</strong>iling ...................................................... 101<br />

5.4 Results .................................................................................................................................. 102<br />

5.4.1 Tissue fixation and staining .............................................................................................. 102<br />

5.4.2 RNA quality and yield from pooled microdissected samples ............................................ 105<br />

5.4.3 RNA amplification and microarray hybridization ............................................................... 106<br />

5.4.4 Performance <strong>of</strong> the established protocol .......................................................................... 110<br />

5.5 Discussion............................................................................................................................. 111<br />

5.6 Appendix ............................................................................................................................... 112<br />

Chapter 6: Manuscript IV ................................................................................................................... 114<br />

6.1 Abstract ................................................................................................................................. 114<br />

6.2 Introduction ........................................................................................................................... 115<br />

6.3 Material and Methods............................................................................................................ 116<br />

6.3.1 Compounds ...................................................................................................................... 116<br />

6.3.2 Animals............................................................................................................................. 116<br />

6.3.3 Sample collection ............................................................................................................. 117<br />

6.3.4 Histopathology.................................................................................................................. 117<br />

6.3.5 Immunohistochemistry...................................................................................................... 117<br />

6.3.6 Laser microdissection and RNA isolation ......................................................................... 118<br />

6.3.7 Microarray data processing and analysis ......................................................................... 118<br />

6.3.8 Statistical analysis ............................................................................................................ 119<br />

6.4 Results .................................................................................................................................. 119<br />

6.4.1 Non-neoplastic pathology ................................................................................................. 119<br />

6.4.2 Cell proliferation................................................................................................................ 120<br />

6.4.3 (Pre-) neoplastic pathology............................................................................................... 121<br />

6.4.4 Gene expression pr<strong>of</strong>iling................................................................................................. 123<br />

VII


6.4.5 Confirmation <strong>of</strong> TORC1 and TORC2 pathway activation in preneoplastic lesions ........... 127<br />

6.5 Discussion............................................................................................................................. 130<br />

6.5.1 Compound-induced tubular damage and regenerative cell proliferation is critical for the<br />

formation but not for the progression <strong>of</strong> preneoplastic lesions......................................................... 130<br />

6.5.2 Time dependent tissue adaptation to carcinogen treatment ............................................. 131<br />

6.5.3 Numbers <strong>of</strong> earliest preneoplastic lesions formed are decisive for later occurrence <strong>of</strong><br />

veritable tumors ............................................................................................................................... 132<br />

6.5.4 The balance <strong>of</strong> TORC1 and TORC2 pathways determines preneoplastic lesion progression<br />

133<br />

6.5.5 Outlook ............................................................................................................................. 134<br />

6.6 Supplemental Material........................................................................................................... 135<br />

6.6.1 Supplementary experimental procedures: Real time PCR ............................................... 135<br />

6.6.2 Supplementary Figures .................................................................................................... 136<br />

Supplemental Tables ....................................................................................................................... 140<br />

Chapter 7: Manuscript V .................................................................................................................... 145<br />

7.1 Abstract ................................................................................................................................. 145<br />

7.2 Introduction ........................................................................................................................... 146<br />

7.3 Material and Methods............................................................................................................ 148<br />

7.3.1 Animals............................................................................................................................. 148<br />

7.3.2 Blood parameters ............................................................................................................. 148<br />

7.3.3 Renal Pathology ............................................................................................................... 149<br />

7.3.4 Cell Proliferation analysis ................................................................................................. 149<br />

7.3.5 Immunohistochemistry...................................................................................................... 149<br />

7.3.6 Statistics ........................................................................................................................... 150<br />

7.4 Results .................................................................................................................................. 150<br />

7.4.1 Wistar rats show widely differing propensity towards body weight gain in response to<br />

prolonged high-fat diet exposure...................................................................................................... 150<br />

7.4.2 Increased levels <strong>of</strong> adipokines and insulin in DIOsens and DIOres rats........................... 151<br />

7.4.3 High-fat diet exposure causes preneoplasia in DIOsens and DIOres rats........................ 152<br />

7.4.4 High-fat diet-induced changes in kidney pathology are independent from α2u-globulin<br />

accumulation.................................................................................................................................... 155<br />

7.4.5 High fat diet increases cell proliferation in proximal tubule <strong>of</strong> DIOsens and DIOres rats.. 155<br />

VIII


7.4.6 Involvement <strong>of</strong> the mTOR pathway .................................................................................. 156<br />

7.5 Discussion............................................................................................................................. 157<br />

7.5.1 Similar renal preneoplasias in DIOsens and DIOres rats suggest a minor role <strong>of</strong> high body<br />

adiposity or its co-morbidities........................................................................................................... 158<br />

7.5.2 High-fat diet exposure does not induce α2u-globulin accumulation................................... 159<br />

7.5.3 mTor-S6K signaling: The link between obesity and kidney cancer................................... 160<br />

7.5.4 Conclusion........................................................................................................................ 161<br />

Chapter 8: Additional Data ................................................................................................................. 162<br />

8.1 Gender Disparity in Early Ochratoxin A and Aristolochic Acid Mediated Renal Toxicity in Shortterm<br />

Assays ......................................................................................................................................... 162<br />

8.2 Ochratoxin A ......................................................................................................................... 162<br />

8.2.1 Cell proliferation (PCNA labeling index) ........................................................................... 162<br />

8.2.2 Renal pathology................................................................................................................ 163<br />

8.2.3 Conclusion........................................................................................................................ 164<br />

8.3 Aristolochic acid .................................................................................................................... 165<br />

8.3.1 Cell proliferation (PCNA labeling index) ........................................................................... 165<br />

8.3.2 Renal pathology................................................................................................................ 165<br />

8.3.3 Conclusion........................................................................................................................ 166<br />

Chapter 9: Overall Discussion and Future Perspectives .................................................................... 167<br />

9.1 Short-term In Vivo Tests to Identify Renal Carcinogens........................................................ 167<br />

9.1.1 Prerequisite I: Short term treatment would result in detectable compound-specific gene<br />

expression changes ......................................................................................................................... 168<br />

9.1.2 Prerequisite II: Genotoxic and non-genotoxic renal carcinogens can be distinguished by<br />

early gene expression pr<strong>of</strong>iles ......................................................................................................... 171<br />

9.1.3 Prerequisite III: Manifestation <strong>of</strong> early compound-induced expression changes in kidney<br />

tumors. 172<br />

9.1.4 Summary Part I................................................................................................................. 174<br />

9.2 Cellular Signaling Cascades in Preneoplastic Lesion Progression ....................................... 175<br />

9.2.1 Pivotal role <strong>of</strong> carcinogen-induced tubular damage, regeneration and adaptation processes<br />

for the development <strong>of</strong> kidney cancer .............................................................................................. 176<br />

9.2.2 Predictive potential <strong>of</strong> preneoplastic lesions as tumor precursors .................................... 180<br />

9.2.3 Summary Part II................................................................................................................ 183<br />

IX


9.3 Obesity as biasing factor for a reliable human risk assessment from rodent bioassays........ 184<br />

9.4 Synopsis................................................................................................................................ 186<br />

Chapter 10: References ....................................................................................................................... 188<br />

Erklärung.................................................................................................................................................. 199<br />

Danksagung ............................................................................................................................................. 200<br />

X


List <strong>of</strong> Figures<br />

Figure 1.1: Overview <strong>of</strong> the morphological structures <strong>of</strong> the renal nephron.. ................................ 5<br />

Figure 1.2: Progression <strong>of</strong> renal lesions........................................................................................ 8<br />

Figure 1.3: Chemical structures <strong>of</strong> AAI and AAII......................................................................... 13<br />

Figure 1.4: Chemical structure <strong>of</strong> OTA........................................................................................ 15<br />

Figure 1.5: Chemical structures <strong>of</strong> cycasin, MAM and methyldiazonium ion............................... 17<br />

Figure 1.6: Composition <strong>of</strong> an Affymetrix chip............................................................................. 23<br />

Figure 2.1: Overview on the experimental design ....................................................................... 32<br />

Figure 3.1: Comparison <strong>of</strong> PCNA S-Phase labelling indices: short term assay ......................... 40<br />

Figure 3.2: PCNA staining patterns <strong>of</strong> control- and OTA-treated Eker rats: short term assay. ... 41<br />

Figure 3.3: AA or OTA induced renal gene expression pr<strong>of</strong>iles in short term assays ................. 42<br />

Figure 3.4: Postulated mechanistic pathways <strong>of</strong> AA and OTA induced toxicity........................... 59<br />

Figure 4.1: Metabolic pathways <strong>of</strong> cycasin and MAM activation ................................................. 75<br />

Figure 4.2: MAMAc induced renal gene expression pr<strong>of</strong>iles in short term assays ...................... 80<br />

Figure 4.3: Representative preneoplastic lesions in MAMAc treated rats ................................... 85<br />

Figure 4.4: Mean number <strong>of</strong> preneoplastic lesions in MAMAc treated Eker rats......................... 86<br />

Figure 4.5: PCNA S-Phase mean labeling indices in MAMAc treated rats.................................. 87<br />

Figure 4.6: Number <strong>of</strong> O6MG adducts detected via LC-MS/MS ................................................. 88<br />

Figure 5.1: Histology <strong>of</strong> H&E stained renal cryo-sections. ........................................................ 103<br />

Figure 5.2: RNA quality in renal cyrosections ........................................................................... 105<br />

Figure 5.3: Comparison <strong>of</strong> total RNA yields after one- or two cycle amplification .................... 107<br />

Figure 5.4: Expression pr<strong>of</strong>iles <strong>of</strong> differentially expressed genes. ........................................... 108<br />

Figure 5.5.: Principal components analysis: One cycle and two-cyce amplification ................... 109<br />

Figure 5.6. Boxplot: Analyis <strong>of</strong> Affymetrix genechip data .......................................................... 110<br />

Figure 6.1. Preneoplastic lesions in AA- and OTA-treated rats: chronic exposure.................... 122<br />

Figure 6.2: Heatmap comparing gene expression changes <strong>of</strong> microdissected lesions ............. 123<br />

Figure 6.3. Postulated mechanisms involved in chemically induced in renal cacinogenesis..... 127<br />

Figure 6.4: mTOR activation in preneoplastic lesions............................................... ............. ...129<br />

Figure 7.1: Adipokine, cytokine, and insulin levels ................................................................... 152<br />

Figure 7.2: Images <strong>of</strong> renal histopathology in DIOsens and DIOres rats .................................. 154<br />

Figure 7.3: PCNA staining in DIOsens and DIOres rats............................................................ 156<br />

Figure 7.4: pS6RP staining in DIOsens and DIOres rats .......................................................... 157<br />

Figure 8.1: PCNA labelling indices in OTA treated Eker and willd type rats.............................. 163<br />

Figure 8.2: Total numbers and incidences <strong>of</strong> bATs in OTA treated rats.................................... 164<br />

Figure 8.3: PCNA labelling indices in AA treated Eker and willd type rats. .............................. 165<br />

Figure 8.4: Total numbers and incidences <strong>of</strong> bATs in AA treated rats ...................................... 166<br />

XI


Figure 9.1: Hypothetical model <strong>of</strong> chemically-induced renal carcinogenesis ........................... 177<br />

Figure 9.2: Gene expression pr<strong>of</strong>iles from short-term treated rats and HT .............................. 179<br />

Figure 9.3: Overview <strong>of</strong> the mTOR pathway ............................................................................ 183<br />

Supplementary Figures:<br />

Supplementary figure 4.1: Real time PCR MGMT...................................................................................... 92<br />

Supplementary figure 6.1: Overview over the experimental design.......................................................... 136<br />

Supplementary figure 6.2: H&E stained paraffin sections: Renal pathology............................................. 137<br />

Supplementary figure 6.3; Immunmohistochemistry pS6RP, pAKT, Foxo1 ........................................... 138<br />

Supplementary figure 6.4: Real time PCR: OGG1 and CAT .................................................................... 139<br />

XII


List <strong>of</strong> Tables<br />

Table 1.1: Classification <strong>of</strong> kidney tumors.................................................................................... 6<br />

Table 1.2: Images <strong>of</strong> frequent cellular abnormalities in rat kidney tumors ................................... 9<br />

Table 3.1: Gene categories <strong>of</strong> genes differentially deregulated by AA ...................................... 45<br />

Table 3.2: Gene categories <strong>of</strong> genes differentially deregulated by OTA.................................... 50<br />

Table 4.1: Genes <strong>of</strong> genes differentially deregulated by MAMAc .............................................. 81<br />

Table 4.2: Non-neoplastic renal pathology in MAMAc treated Eker r......................................... 84<br />

Table 5.1: Influence <strong>of</strong> tissue preservation on RNA quality...................................................... 102<br />

Table 5.2: Influence <strong>of</strong> long-term storage on RNA quality and yield ........................................ 103<br />

Table 5.3: RNA quality and yields from pooled microdissected samples................................. 106<br />

Table 6.1: Comparison <strong>of</strong> BrdU S-Phase labeling indices: Chronic treatment ......................... 120<br />

Table 6.2: Numbers and categories <strong>of</strong> deregulated genes in preneoplastic lesions ................ 125<br />

Table 7.1: Overview <strong>of</strong> antibodies and modifications to the manufacture’s protocols .............. 149<br />

Table 7.2: Life-history traits <strong>of</strong> chow-fed rats, DIOres and DIOsens rats ................................. 151<br />

Table 7.3: Histopathology in chow-fed, DIOres, and DIOsens rats.......................................... 153<br />

Table 9.1: Pathways involved in AA-, OTA- or MAMAc-induced renal carcinogenicity ............ 170<br />

Table 9.2: Genes and pathways linked to the six hallmarks <strong>of</strong> cancer......................................181<br />

Supplementary Tables:<br />

Supplementary table 3.1: Numbers and categories <strong>of</strong> genes deregulated by AA or OTA.......................... 63<br />

Supplementary table 3.2: Incidence and numbers <strong>of</strong> AA and OTA induced renal lesions.......................... 65<br />

Supplementary table 3.3: Non-neoplastic pathology <strong>of</strong> AA or OTA treated Eker rats. ............................... 66<br />

Supplementary table 3.4: Non-neoplastic pathology <strong>of</strong> AA or OTA treated wild type rats. ........................ 70<br />

Supplementary table 4.1: Non-neoplastic pathology <strong>of</strong> MAMAc treated rats ............................................. 93<br />

Supplementary table 4.2: Incidence and numbers <strong>of</strong> MAMAc induced renal lesions. ................................ 94<br />

Supplementary table 6.1: Non-neoplastic renal pathology: 6months treatment ....................................... 140<br />

Supplementary table 6.2: (Pre-)neoplastic pathology: 6 months treatment ............................................. 141<br />

Supplementary table 6.3: Genes differentially deregulated in preneoplastic lesions ............................... 142<br />

XIII


Abbreviations<br />

α2u alpha 2 urine-globulin<br />

AA aristolochic acid<br />

AAN aristolochic acid nephropathy<br />

bAT basophilic atypical tubule<br />

bAH basophilic atypical hyperplasia<br />

BEN Balkan endemic nephropathy<br />

bw body weight<br />

CPN chronic progressive nephropathy<br />

DIO diet induced obesity<br />

ECM extracellular matrix<br />

EK Eker<br />

EST expressed sequence tag<br />

HT healthy tubule<br />

IARC International Agency for Research on <strong>Cancer</strong><br />

ICH International Conference on Harmonization<br />

i.p. intraperitoneal<br />

LC/MS Liquid Chromatography/ Mass spectrometry<br />

LOH loss <strong>of</strong> heterozygosity<br />

MAM mehylazoxymethanol<br />

MAMAc mehylazoxymethanol acetate<br />

MTD maximum tolerated dose<br />

mTOR Mammalian target <strong>of</strong> rapamycin<br />

NNM N-nitrosodimethylamine<br />

NOAEL no observable adverse effect level<br />

NTP National Toxicology Program<br />

OTA ochratoxin A<br />

ROS reactive oxygen species<br />

TSC tuberous sclerosis complex<br />

UT urothelial tumors<br />

WHO World Health Organization<br />

WT wild type<br />

XIV


Journal Articles<br />

Publications and Presentations<br />

• Kerstin Stemmer, Heidrun Ellinger-Ziegelbauer, Hans-Juergen Ahr and Daniel R.<br />

Dietrich: Carcinogen Specific Gene Expression Pr<strong>of</strong>iles in Short-Term Treated Eker<br />

and Wild Type Rats Indicative for Pathways Involved in Renal Tumorigenesis.<br />

<strong>Cancer</strong> Research (2007) May 1;67(9):4052-68<br />

• Kerstin Stemmer, Heidrun Ellinger-Ziegelbauer, Kerstin Lotz, Hans-J. Ahr and<br />

Daniel R. Dietrich: Establishment <strong>of</strong> a Protocol for the Analysis <strong>of</strong> Laser<br />

Microdissected Rat <strong>Kidney</strong> Samples on Affymetrix Genechips. Toxicology and<br />

Applied Pharmacology, 217 (2006) 134-142.<br />

• Kerstin Stemmer, Heidrun Ellinger-Ziegelbauer, Hans-Juergen Ahr and Daniel R.<br />

Dietrich: Earliest preneoplastic renal lesions provide an in-depth understanding <strong>of</strong><br />

renal carcinogenesis (American Journal <strong>of</strong> Pathology, submitted)<br />

• Kerstin Stemmer, Heidrun Ellinger-Ziegelbauer, Katja Strauch, Leonard Collins,<br />

James A. Swenberg, Hans-Juergen Ahr and Daniel R. Dietrich: Renal Carcinogenic<br />

Response <strong>of</strong> Eker Rats to Low Doses <strong>of</strong> Methylazoxymethanol Acetate (Mutation<br />

Research, submitted)<br />

• Kerstin Stemmer, Diego Perez-Tilve, Daniel R. Dietrich, Matthias H. Tschöp and<br />

Paul T. Pfluger: High-fat diet exposure is associated with renal carcinogenesis in rats<br />

(manuscript to be submitted)<br />

Poster Presentations<br />

• K. Stemmer, P.T. Pfluger, D. Perez-Tilve, M.H. Tschöp and D.R. Dietrich: Long-term<br />

exposure to high-fat diet leads to renal pathology in rats: SOT 47th Annual Meeting,<br />

2008, Seattle, USA<br />

• D.R. Dietrich and K. Stemmer: Gender disparity in early ochratoxin A mediated<br />

toxicity: SOT 47th Annual Meeting, 2008, Seattle, USA<br />

XV


• K. Stemmer and D.R. Dietrich: Characterisation <strong>of</strong> Renal Preneoplastic Lesions in<br />

Carcinogen Treated and Untreated Eker Rats: International Congress <strong>of</strong> Toxicology,<br />

Toxicology Discovery Serving Society, July 2007 Montréal, Canada<br />

• K. Stemmer, H. Ellinger-Ziegelbauer, T. Lampertsdoerfer, K. Lotz, H.J. Ahr and D.R.<br />

Dietrich: Expression Pr<strong>of</strong>iles Induced By Renal Carcinogens in Eker Rats Compared<br />

to Wildtype Rats. SOT 45th Annual Meeting, 2006, San Diego, USA<br />

• K. Stemmer, J. Haehnlein, H. Ellinger-Ziegelbauer, H.J. Ahr and D.R. Dietrich:<br />

Identification <strong>of</strong> Potential Markers for an In Vitro Test System <strong>of</strong> Renal<br />

Carcinogenesis. 5th World Congress on Alternatives & Animal Use in the Life<br />

Sciences, 2005, Berlin, Germany<br />

• K. Stemmer, H. Ellinger-Ziegelbauer, T. Lampertsdoerfer, M. Thiel, H.J. Ahr and<br />

D.R. Dietrich: Ochratoxin A-Induced Gene Expression Deregulations in the <strong>Kidney</strong> <strong>of</strong><br />

Eker Rats Analyzed on Affymetrix Chips. SOT 44th Annual Meeting, 2005, New<br />

Orleans, USA<br />

• K. Stemmer, H. Ellinger-Ziegelbauer, T. Lampertsdoerfer, M. Thiel, H.J. Ahr and<br />

D.R. Dietrich: Expression Pr<strong>of</strong>iles Induced by Renal Carcinogens in Rat <strong>Kidney</strong><br />

Analysed on Affymetrix Chips. Joint Speciality Symposium on Renal Toxicology and<br />

Toxicologic Pathology, 2004, Lindau, Germany<br />

• J. Haehnlein, K. Stemmer, H. Ellinger-Ziegelbauer, S. Michel-Kaulmann, H.J. Ahr<br />

and D.R. Dietrich: Cytotoxic Effects <strong>of</strong> Aristolochic Acid (AA) on NRK-52E and<br />

Primary Rat <strong>Kidney</strong> Cells and Associated Changes in the Expression Pattern <strong>of</strong> p21<br />

and MGMT. Joint Speciality Symposium on Renal Toxicology and Toxicologic<br />

Pathology, 2004, Lindau, Germany<br />

XVI


Oral presentations<br />

• Früherkennung Nieren-kanzerogener Substanzen. Projektforum Biotechnologie<br />

German Federal Ministry <strong>of</strong> Research and Education (BMBF), 10.10.2007,<br />

BIOTECHNIKA Hannover, Germany.<br />

• The Use <strong>of</strong> Laser Microdissection and Pressure Catapulting to Study Gene<br />

Expression in Renal Preneoplastic Lesions. P.A.L.M MicroBeam Workshop:<br />

16.11.2006, P.A.L.M Microlaser Technologies AG, Bernried, Germany.<br />

• Gene Expression Patterns <strong>of</strong> Different Stages <strong>of</strong> Renal Pre-Neoplasia: Biolago<br />

Symposium: Science Meets Economy: 13.10.2006, University <strong>of</strong> Konstanz,<br />

Germany.<br />

Funding and Awards<br />

• Three months research visit at the University <strong>of</strong> Tel Aviv in Israel. Funded by the<br />

Young Scientist Exchange Program <strong>of</strong> the German Federal Ministry <strong>of</strong> Research and<br />

Education (BMBF) and Israel’s Ministry <strong>of</strong> Science, Culture and Sports (MOST),<br />

2008.<br />

• Three months PhD-scholarship <strong>of</strong> the Office <strong>of</strong> Equal Opportunity and Women‘s<br />

Affairs, University <strong>of</strong> Konstanz, 2008.<br />

• Congress Grant by the German Research Foundation (DFG), 2008<br />

• Research Grant: <strong>Molecular</strong> Analysis <strong>of</strong> the Nephrotoxic Action <strong>of</strong> the Mycotoxin<br />

Ochratoxin A. Environment and Living Foundation, University <strong>of</strong> Konstanz, 2006<br />

• Three months PhD-scholarship <strong>of</strong> the Senate Committee for Research (AFF) <strong>of</strong> the<br />

University <strong>of</strong> Konstanz, 2006.<br />

• Travel Award <strong>of</strong> the Society <strong>of</strong> Toxicology SOT, 2006<br />

• Finalist for the CVSS (Comparative & Veterinary Speciality Section) Student Poster<br />

Award, SOT 45th Annual Meeting in San Diego, USA, 2006<br />

XVII


Zusammenfassung<br />

Hintergrund: Die Mehrheit aller Krebserkrankungen, darunter auch Nierenkrebs, werden<br />

durch Fremdst<strong>of</strong>fe verursacht. Zur Untersuchung des möglichen kanzerogenen Potentials<br />

von Fremdst<strong>of</strong>fen sind zweijährige Kanzerogenesestudien in Nagetieren vom Gesetzgeber<br />

vorgeschrieben. In diesen Tests werden nach zweijähriger Exposition mit der jeweiligen<br />

Testsubstanz die Tumorinzidenz und weitere pathologische Effekte untersucht und<br />

anschliessend eine Risikoabschätzung für den Menschen erstellt. Derartige Studien sind<br />

jedoch äußerst zeitaufwendig und teuer. Hinzu kommt ein hoher Tierverbrauch sowie ein<br />

hoher Bedarf an Testsubstanzen für die zweijährige Dosierung. Erschwerend kommt hinzu,<br />

dass die Ergebnisse dieser Langzeit-Studien <strong>of</strong>t durch unspezifische Hochdosiseffekte<br />

oder durch eine altersabhängige Pathologie verfälscht werden. Zudem können Nagetierspezifische<br />

Effekte auftreten, die für eine Abschätzung der potentiellen Risiken für den<br />

Menschen nur wenig relevant sind, Umgekehrt vernachlässigen diese Studien jedoch<br />

andere Krebsrisik<strong>of</strong>aktoren, etwa eine hochkalorische Diät oder Fettleibigeit, die die<br />

kanzerogene Wirkung eines Fremdst<strong>of</strong>fes verstärken können. Die Entwicklung neuer,<br />

hochsensitiver sowie robuster Kurzzeit-In-Vivo-Testsysteme könnte bereits nach kurzen<br />

Expositionsdauern Aussagen über das kanzerogene Potential sowie den<br />

Wirkungsmechanismus einer Testsubstanz liefern, und zu einer verbesserte<br />

Risikoabschätzung für den Menschen beitragen.<br />

Ziele: Im Rahmen dieser Doktorarbeit sollte untersucht werden, ob Nieren-kanzerogene<br />

Substanzen in Kurzeit-In-Vivo-Studien durch sensitive Microarray-Transkriptomanalysen<br />

identifiziert werden können. Des weiteren sollten molekulare Mechanismen bei der<br />

Entwicklung von Fremdst<strong>of</strong>f-induzierten Nierentumoren charakterisiert werden. Schließlich<br />

sollten die Einflüsse einer fetthaltigen Diät sowie erhöhter Fettleibigkeit auf frühe Stadien<br />

von Nierenkrebs in der Ratte bestimmt werden.<br />

Experimenteller Aufbau: I) Wildtyp Ratten sowie Eker Ratten mit einer Mutation im TSC2<br />

Tumorsuppressor Gen wurden täglich mit den genotoxischen Nierenkanzerogenen<br />

Aristolochia Säure (AA), Methylazoxymethanol Azetat (MAMAc), oder dem nicht<br />

genotoxischen Nierenkanzerogen Ochratoxin A (OTA) gavagiert. Nach 1, 3, 7 und 14<br />

Tagen wurden von Nierenkortex-Homogenaten aller behandelten und Kontroll-Tiere<br />

Genexpressionspr<strong>of</strong>ile mittels Microarrays erstellt. Paraffinschnitte wurden zur<br />

Untersuchung von Histopathologie und Zellproliferation verwendet. II) Eker Ratten wurden<br />

XVIII


mit AA, MAMAc und OTA über 3 und 6 Monate gavagiert und histopathologische<br />

Veränderungen und Zellproliferationraten bestimmt. Parallel hierzu wurde ein Protokoll<br />

entwickelt, welches erstmals reproduzierbare und verlässliche Genexpressionsanaysen<br />

von mikrodissektierten Nierentubuli ermöglichte: Nach diesem Protokoll wurden<br />

Transkriptionsanalysen aus frühen Vortumorstadien (Prä-neoplasien) sowie aus gesunden<br />

Nierentubuli AA- und OTA- und Vehikel-behandelter Eker Ratten erstellt. Die zentralen<br />

Ergebnisse dieser Genexpressionsanalysen wurden mittels immunhistochemischen<br />

Färbungen verifiziert. III) Adverse Effekte auf Nierenpathologie und zelluläre Signalwege<br />

wurden in zwei Subpopulationen von Wistar-Ratten mit hoher Sensitivitaet bzw. Resistenz<br />

gegenüber nahrungsinduzierter Adipositas nach 11 Monaten Exposition mit fetthaltiger<br />

(45%) Diät, sowie in Wistar-Ratten nach Exposition mit fettarmer (6%) Diät untersucht.<br />

Ergebnisse & Diskussion: Histopatholgische Untersuchungen sowie v.a. die Auswertung<br />

der Genexpressionspr<strong>of</strong>ile aus Nierenkortex-Homogenaten zeigten, dass früheste<br />

substanzspezifische Effekte Nieren-kanzerogener Substanzen bereits nach 1- bis 14tägiger<br />

Exposition mit AA, MAMAc bzw. OTA erkennbar sind. Des weiteren erlaubten<br />

besagte Genexpressionspr<strong>of</strong>ile auch die Unterscheidung von genotoxischen und nichtgenotoxischen<br />

Substanzen. Diese Veränderungen der Genexpression nach 1-14 Tagen<br />

Exposition mit AA, MAMA oder OTA waren zudem prediktiv für die substanzspezifische<br />

Inzidenz und Zahl von prä-neoplastischen Läsionen in Eker Ratten nach 3- bzw. 6monatiger<br />

Exposition.<br />

Genexpressionspr<strong>of</strong>ile aus mikrodissektierten präneoplastischen Läsionen von Eker Ratten<br />

nach 6-monatiger Exposition mit AA, OTA oder dem entsprechenden Vehikel zeigten im<br />

Vergleich zu mikrodissektierten gesunden Tubuli von Vehikel behandelten Ratten eine<br />

stark erhöhte Deregulation zahlreicher Gene. Interessanterweise ähnelten sich die<br />

Expressionveränderungen in den verschiedenen Stadien der Läsionen und nach<br />

Behandlung mit den verschiedenen Kanzerogenen stark. Des weiteren konnten nur<br />

geringfügige Genexpressionveränderungen in gesunden Tubuli AA- und OTA-behandelter<br />

Ratten gemessen werden, was auf deren Insensitivität gegenüber kanzerogenen<br />

Substanzen hindeutetete. Diese Daten weisen darauf hin, dass die Anzahl der<br />

präneoplastischen Läsionen, die während einer begrenzten kritischen Phase der<br />

Substanzbehandlung entstehen, ausschlaggebend für die Inzidenz und Anzahl von<br />

Tumoren am Ende der zweijährigen Kanzerogenese Studie sein könnte. Die klonale<br />

Expansion dieser Läsionen ist demgegenüber eher ein substanzunabhängiger Prozess, der<br />

möglicherweise durch eine gestörte Feedback-Inhibitierung des mTOR Signalwegs<br />

verstärkt wird.<br />

XIX


Im letzten Teil der Dissertation konnte gezeigt werden, dass eine stark fetthaltige Diät eine<br />

deutlich schädliche Wirkung auf Rattennieren ausübt. Hierbei stellte sich jedoch heraus,<br />

dass der Grad der Adipositas nur eine untergeordnete Rolle einnimmt im Vergleich zu den<br />

direkten adversen Effekten der ueber die Nahrung aufgenommenen Lipide.<br />

Immunohistochemische Färbungen der Nieren wiesen zudem auf eine bedeutende Rolle<br />

des mTOR Signalweges bei der Lipid-induzierten preneoplastischen Pathologie der Niere.<br />

Fazit: Kurzzeit-In-Vivo-Studien in Kombination mit Microarrays erlauben die<br />

Charakterisierung von frühesten molekularen Vorgängen bei der Entstehung von<br />

Fremdst<strong>of</strong>f-induzierten Nierentumoren. Des weiteren erfüllen sie alle wichtigen<br />

Vorraussetzungen für eine verlässliche Identifizierung von Nieren-kanzerogenene<br />

Substanzen, nämlich eine hohe Sensitivität, Spezifität und Prediktivität. Die Ergebnisse<br />

dieser Arbeit zeigen, dass unter Verwendung neuer und sensitiver Detektionstechniken<br />

z,B. Microarrays, zweijährige Kanzerogenesestudien deutlich verkürzt werden könnten. Auf<br />

Basis der hier gezeigten Erkenntnisse erscheint es deshalb durchaus möglich, neue<br />

verbesserte Kanzerogenese-In-Vivo-Tests zu entwickeln, die zudem zu einer Reduktion<br />

der Tierzahl führen und weiterhin molekulare Grundlagen schaffen, die zukünftig zur<br />

Schaffung tierversuchsfreier Methoden dienen könnten.<br />

XX


Summary<br />

Background: The vast majority <strong>of</strong> all human cancers, including kidney cancer, are caused<br />

by xenobiotica. Two-year rodent bioassays play the central role in evaluating the<br />

carcinogenicity <strong>of</strong> chemicals and are mainly based on a post-mortem evaluation <strong>of</strong> tumor<br />

incidences and other pathological changes after two years <strong>of</strong> exposure. However, such<br />

lifetime bioassays are time-consuming and costly, require large numbers <strong>of</strong> animals and a<br />

high amount <strong>of</strong> compound for continuous dosing over the entire period <strong>of</strong> the assay. In<br />

addition, widespread age-related pathologies, unspecific high-dose effects, or rodentspecific<br />

carcinogenic effects with little or no relevance to humans can confound the<br />

outcome <strong>of</strong> these studies. Furthermore, they do not account for possible additive effects <strong>of</strong><br />

other cancer risk factors, such as diet-induced obesity. Therefore, interest in developing<br />

short, sensitive but robust, and mechanistically based in vivo studies has grown, allowing a<br />

more reliable human risk assessment.<br />

Aims: The overall aim <strong>of</strong> this thesis was to elucidate whether short-term in vivo assays in<br />

combination with microarray technology can be used to identify renal carcinogens. In<br />

addition, a more detailed understanding <strong>of</strong> potential additive effects <strong>of</strong> high fat diet<br />

exposure, and molecular mechanisms underlying chemically induced renal carcinogenesis,<br />

should be obtained.<br />

Experimental Setup: I) TSC2-mutant Eker and wild type rats were gavaged daily with the<br />

genotoxic renal carcinogens aristolochic acid (AA) and methylazoxymethanol acetate<br />

(MAMAc), or with the non-genotoxic ochratoxin A (OTA), respectively. Subsequent 1, 3, 7<br />

and 14 days <strong>of</strong> exposure, gene-expression pr<strong>of</strong>iles from kidney cortex homogenates,<br />

histopathology, and cell proliferation were assessed. II) Eker rats were gavaged with AA,<br />

MAMAc and OTA for 3 or 6 months. Subsequently, renal histopathology and cell<br />

proliferation were evaluated. In addition, a novel protocol for reproducible and reliable gene<br />

expression analysis from laser-microdissected pre-neoplastic renal lesions was<br />

established, and gene expression pr<strong>of</strong>iles <strong>of</strong> different stages <strong>of</strong> preneoplastic lesions and<br />

healthy tubules were examined from 6 months AA-, OTA- and vehicle-treated male Eker<br />

rats. Key findings from transcriptome analyses were verified by immunohistochemistry. III)<br />

The respective impacts <strong>of</strong> dietary lipids and high body adiposity on renal pathology and<br />

pathways involved in renal cancer were delineated in diet-induced obesity-sensitive and<br />

XXI


diet-induced obesity-resistant subpopulations <strong>of</strong> male Wistar rats vs. chow-fed controls,<br />

subsequent to 11 months <strong>of</strong> high fat diet or standard chow exposure, respectively.<br />

Results & Discussion: Renal histopathology and gene expression pr<strong>of</strong>iling <strong>of</strong> kidney<br />

homogenates <strong>of</strong> short-term AA-, MAMAc-, and OTA-treated rats revealed that microarray<br />

technology is a sensitive tool to study earliest events <strong>of</strong> renal carcinogenesis. In addition,<br />

gene expression pr<strong>of</strong>iles allowed dissecting genotoxic from non-genotoxic modes <strong>of</strong> action.<br />

Most importantly, early gene expression changes after short-term exposure were predictive<br />

for the incidence and number <strong>of</strong> preneoplastic lesions after 3 or 6 months <strong>of</strong> exposure.<br />

Gene expression pr<strong>of</strong>iles from microdissected preneoplastic lesions <strong>of</strong> 6-month AA-, OTAor<br />

control rats revealed marked gene deregulations, when compared to healthy tissue <strong>of</strong><br />

control animals. Notably, gene expression changes were similar in different lesion<br />

progression stages and after treatment with genotoxic or non-genotoxic carcinogens.<br />

Furthermore, gene expression pr<strong>of</strong>iles <strong>of</strong> microdissected healthy tubules <strong>of</strong> AA- and OTAtreated<br />

rats were only marginally changed, suggesting a low compound-sensitivity. These<br />

findings suggested that the number <strong>of</strong> preneoplastic lesions initially formed during a critical<br />

period <strong>of</strong> exposure may primarily drive the incidence and number <strong>of</strong> solid tumors. In<br />

contrast, clonal expansion <strong>of</strong> these lesions may be compound-independent and instead be<br />

driven by a disturbed feedback inhibition <strong>of</strong> the mTOR pathway.<br />

Last, high fat diet exposure <strong>of</strong> male Wistar rats revealed a clear adverse effect <strong>of</strong> dietary<br />

lipids, and a minor role <strong>of</strong> the degree <strong>of</strong> body adiposity, on renal morphology. In addition,<br />

immunohistochemical analyses suggested a major role <strong>of</strong> the mTOR pathway in dietary<br />

lipid-induced pre-neoplastic pathology.<br />

Conclusion: Microarray-based short-term in vivo assays not only help to delineate<br />

molecular events important for renal cancer, but also fulfill the most important prerequisites<br />

for reliable carcinogenicity testing, i.e. high sensitivity, high specificity, and high predictivity.<br />

Thus, the duration <strong>of</strong> the standard two-year rodent bioassay can be reduced to a shorter<br />

period <strong>of</strong> time by using novel and sensitive methodology such as microarrays. Ultimately,<br />

this thesis provides encouraging first results that may help to reduce, refine and one day<br />

even replace animal experiments.<br />

XXII


1.1 <strong>Kidney</strong> <strong>Cancer</strong><br />

Chapter 1: Introduction<br />

<strong>Kidney</strong> cancer accounts for 2% <strong>of</strong> all malignancies worldwide, with approximately 210.000<br />

additional cases per year. Because <strong>of</strong> its high mortality and increasing incidence in most<br />

countries, it is considered to be one <strong>of</strong> the most important urologic cancers. The world’s<br />

highest incidence <strong>of</strong> kidney cancer is observed in Eastern European countries (Czech<br />

Republic, Estonia and Hungary). Germany has the 6th highest incidence with 13.2 cases<br />

and 6.3 mortalities per 100.000 in men and 5.9 cases and 2.9 mortalities per 100.000 in<br />

women (IARC 2002c). <strong>Kidney</strong> tumors usually do not cause signs or symptoms and <strong>of</strong>ten<br />

remain unrecognized in their early and treatable stages. Advanced stages and metastases<br />

are therefore more common with kidney cancer than with many other cancer types.<br />

Secondary tumors <strong>of</strong> the kidney are typically found in the lung, s<strong>of</strong>t tissue, bone, liver,<br />

cutaneous sites and the central nervous system (Corgna et al. 2007).<br />

1.1.1 Risk Factors for <strong>Kidney</strong> <strong>Cancer</strong><br />

The majority <strong>of</strong> human kidney tumors occur sporadically and only 1-4% <strong>of</strong> all kidney cancer<br />

cases are caused by an inherited predisposition (Pavlovich and Schmidt 2004). Sporadic<br />

tumors <strong>of</strong> the kidneys occur more frequently in men than in women (McLaughlin and<br />

Lipworth 2000). The underlying causes for the sex-specific differences are largely unknown,<br />

but may be due to differences in metabolism or hormonal balance. In both sexes kidney<br />

cancer increases with age and more than 75% <strong>of</strong> all cases occur over the age <strong>of</strong> 60.<br />

Besides sex and age, epidemiologic studies point to several other risk factors for renal<br />

cancer, including smoking, prolonged dialysis, hypertension, or the use <strong>of</strong> diuretics and<br />

other anti-hypertensive medication (McLaughlin and Lipworth 2000). In addition, 1/4 <strong>of</strong> all<br />

human kidney cancers were attributed to excess body weight (Mellemgaard et al. 1995).<br />

However, the underlying causal mechanisms that link the respective risk factors to kidney<br />

cancer still remain elusive.<br />

1


Chapter 1: Introduction<br />

Due to their predominant role in excretion, the kidneys are highly vulnerable to a variety <strong>of</strong><br />

circulating toxicants. Approximately one third <strong>of</strong> the plasma water reaching the kidney is<br />

filtered by the glomeruli and 98-99% <strong>of</strong> that filtered plasma water is reabsorbed from the<br />

tubular lumen (Casarett and Doull 1986). Consequently, any drug or chemical that is<br />

present in non-toxic concentrations in the blood can reach the renal epithelial cells in much<br />

higher concentrations after being concentrated with the urine and after being reabsorbed<br />

into the cells.<br />

1.1.2 Genetic and Epigenetic Causes <strong>of</strong> <strong>Cancer</strong><br />

I: Tumor suppressor genes and oncogenes<br />

<strong>Cancer</strong> is usually caused by mutations that are typically found in two classes <strong>of</strong> genes:<br />

<strong>Cancer</strong>-promoting oncogenes and cancer-preventing tumor suppressor genes. Gain <strong>of</strong><br />

function mutations in oncogenes, which are <strong>of</strong>ten involved in signal transduction and<br />

execution <strong>of</strong> mitogenic signals, result in a hyperactive growth and proliferation <strong>of</strong> cells<br />

(Todd and Wong 1999). Loss <strong>of</strong> function mutations in tumor suppressor genes generally<br />

result in dysregulation <strong>of</strong> the cell cycle and DNA-replication, the inhibition <strong>of</strong> programmed<br />

cell death (apoptosis) (Malumbres and Barbacid 2001), or disturbed interaction <strong>of</strong> tumor<br />

cells with protective cells <strong>of</strong> the immune system (immunoresistance) (Parsa et al. 2007).<br />

In 1975, Alfred Knudson and colleagues proposed the two-hit hypothesis, stating that both<br />

copies (alleles) <strong>of</strong> a tumor suppressor gene must be lost or mutated for cancer to occur<br />

(Knudson 1975). Although widely accepted, certain exceptions to the 'two hit' rule were<br />

found. For instance, tumor suppressor genes like p27Kip1 might exhibit haploinsufficiency<br />

after a mutation <strong>of</strong> a single allele, when the remaining functional allele does not translate<br />

into enough protein to preserve a wild-type phenotype (Fero et al. 1998). For oncogenes,<br />

gain <strong>of</strong> function mutations in one allele are usually enough to trigger cancer development<br />

(Todd and Wong 1999).<br />

Besides mutations in tumor suppressor genes and oncogenes, tumorigenesis can be<br />

supported by any event that accelerates the spontaneous rate <strong>of</strong> genetic alterations in the<br />

cell. For instance, hereditary or spontaneous damage in the cellular DNA damage repair<br />

system may favor the accumulation <strong>of</strong> additional mutations, and genetic or genomic<br />

instability <strong>of</strong> cells (Schmutte and Fishel 1999; Jefford and Irminger-Finger 2006).<br />

2


II: Epigenetic alterations<br />

Chapter 1: Introduction<br />

Most recently, several studies pointed towards a critical role <strong>of</strong> reversible epigenetic<br />

alterations in carcinogenesis (Baylin and Herman 2000, Sparmann, 2006, Vucic, 2008).<br />

Epigenetic regulation is generally organized at the level <strong>of</strong> DNA (post-replicative<br />

methylation), RNA (RNA interference), and protein (histone modifications and polycomb<br />

group protein complexes) (Momparler 2003).<br />

Abnormal patterns <strong>of</strong> DNA methylation in cancer cells have been recognized for more than<br />

20 years and global hypomethylation and region-specific hypermethylation are frequent<br />

events in tumor cells (Baylin and Herman 2000). The biological significance <strong>of</strong> DNA<br />

hypomethylation in cancer is less understood. Hypermethylation mainly occurs at CpG<br />

islands within the promoter region <strong>of</strong> a gene, resulting in its suppressed transcription. The<br />

current model suggests that promoter hypermethylation and transcriptional silencing <strong>of</strong><br />

tumor suppressor or DNA repair genes and mutations within these genes occur at least<br />

with a comparable frequency.<br />

Besides DNA methylation, alteration in histone acetylation plays another important role in<br />

the regulation <strong>of</strong> gene expression. Inactive chromatin mediated by histone deacetylation<br />

was demonstrated as one critical component in the silencing <strong>of</strong> tumor suppressor genes.<br />

Different histone deacetylase inhibitors are currently in clinical trials for the treatment <strong>of</strong><br />

cancer (Martinez-Iglesias et al. 2008).<br />

Although still much has to be learned, epigenetic regulation is already assumed to be<br />

equally important for the onset <strong>of</strong> cancer as genetic alterations. The interplay between<br />

genetic and epigenetic changes during tumor progression is thus becoming a major focus<br />

<strong>of</strong> cancer research.<br />

1.1.3 Models <strong>of</strong> Carcinogenesis<br />

I: Multistage model <strong>of</strong> carcinogenesis<br />

The process <strong>of</strong> carcinogenesis is widely accepted as a multistage process, with a sequence<br />

<strong>of</strong> multiple genetic events preceding the onset <strong>of</strong> cancer (Armitage and Doll 1954; Barrett<br />

and Wiseman 1987). The process is based on genetic alterations that have to be fixed<br />

during DNA replication, leading to growth advantage <strong>of</strong> the initiated cell (tumor initiation).<br />

Clonal expansion <strong>of</strong> these cells may lead to the development <strong>of</strong> pre-neoplastic foci with<br />

<strong>characteristics</strong> that favor genetic instability (tumor promotion). Further accumulation <strong>of</strong><br />

3


Chapter 1: Introduction<br />

additional mutations in the fast-growing cells can result in the development <strong>of</strong> a malignant<br />

phenotype that enables the cells to invade the surrounding tissue (tumor progression).<br />

Such a genetic multistage model has been described by Fearon and Vogelstein for<br />

colorectal cancer, where a linear sequence <strong>of</strong> specific genetic events can be correlated with<br />

evolving pathophysiological stages <strong>of</strong> colon carcinogenesis (Fearon and Vogelstein 1990).<br />

Ten years later the linear model <strong>of</strong> multistage carcinogenesis was elaborated by Hanahan<br />

and Weinberg (Hanahan and Weinberg 2000), who described six essential changes in cell<br />

physiology that define the development <strong>of</strong> malignant tumors: (1) self-sufficiency in growth<br />

signals, (2) insensitivity to growth inhibitory signals, (3) evasion <strong>of</strong> apoptosis, (4) limitless<br />

replicative potential, (5) sustained angiogenesis and (6) tissue invasion and metastasis. In<br />

contrast to the Fearon-Vogelstein model, the Hanahan–Weinberg model does not depend<br />

on the sequential accumulation <strong>of</strong> specific mutations for a distinct tumor type but instead<br />

depends on a set <strong>of</strong> functional changes that can result from many different genetic or<br />

epigenetic alterations, independent <strong>of</strong> cell-type, species and aetiology.<br />

II: <strong>Cancer</strong> stem cell model<br />

During recent years, the stem cell model <strong>of</strong> carcinogenesis gained major attention. The<br />

American Association for <strong>Cancer</strong> Research Stem Cell Workshop defined a cancer stem cell<br />

as a cell within a tumor that possesses the capacity to self-renew. Thus, heterogeneous<br />

lineages <strong>of</strong> cancer cells that comprise the tumor may originate from one cancer stem cell.<br />

In this model cancer cells may arise from tissue stem cells that have acquired mutations<br />

that render them cancerous. <strong>Cancer</strong> cells may also arise from differentiated progenitor cells<br />

that "re-acquired" stem cell like properties due to mutations (Clarke et al. 2006). <strong>Cancer</strong><br />

stem cells were originally identified in leukemia (Bonnet and Dick 1997) and later in solid<br />

brain (Singh et al. 2003), breast (Al-Hajj et al. 2003), and colon tumors (O'Brien et al.<br />

2007). However, to date cancer stem cells have not been detected in kidney tumors.<br />

1.1.4 Renal Tumor Pathology<br />

Renal toxicology and pathology are closely related to the kidney’s complex functional<br />

anatomy, with the cortex forming the outer part enclosing the renal medulla and papilla<br />

(Figure 1.1). The functional unit <strong>of</strong> the kidney is the nephron, consisting <strong>of</strong> the glomerulus,<br />

proximal tubule, loop <strong>of</strong> Henle, distal tubule and collecting duct. The proximal tubule is<br />

furthermore dissected into three segments: The tubulus contortus proximalis, consisting <strong>of</strong><br />

the post-glomerular portion (P1), and the more distal portion (P2) <strong>of</strong> the proximal tubule,<br />

4


Chapter 1: Introduction<br />

containing an active endocytosis/lysosomal apparatus, thus representing the site <strong>of</strong> injury<br />

related to lysosomal overload, as well as to protein-bound toxic moieties (Crist<strong>of</strong>ori et al.<br />

2007). The following straight portion <strong>of</strong> the P3 segment (tubulus rectus proximalis)<br />

represents the most susceptible site <strong>of</strong> injury via metabolic activation <strong>of</strong> xenobiotica and via<br />

their transporter-associated accumulation (Crist<strong>of</strong>ori et al. 2007).<br />

Figure 1.1: Overview <strong>of</strong> the morphological structures <strong>of</strong> the renal nephron. Source:<br />

http://de.wikipedia.org/wiki/Nephron.<br />

I: <strong>Kidney</strong> Tumors in Humans<br />

The majority <strong>of</strong> kidney tumors in humans (80-85%) are renal cell carcinoma (RCC),<br />

originating from the renal parenchyma. The remaining 15-20% are mainly transitional cell<br />

carcinoma <strong>of</strong> the renal pelvis, while other neoplasms, e.g. angiomyolipoma or<br />

nephroblastoma (Wilms' Tumor), are rare (Motzer et al. 1996).<br />

RCC are not a single entity but comprise a heterogeneous group <strong>of</strong> tumors that range from<br />

entirely benign to aggressively malignant. They were sub-classified according to their<br />

histological <strong>characteristics</strong> into clear cell RCC (70%), papillary RCC (15%), chromophobe<br />

RCC (10%), oncocytoma (5%) and collecting duct carcinoma (


Chapter 1: Introduction<br />

to correlate with distinct histological sub-types in sporadic RCC, which allowed a more indepth<br />

tumor classification and improved diagnostics (for further information see table 1.1)<br />

(Kovacs et al. 1997; Pavlovich and Schmidt 2004).<br />

Table 1.1: Classification <strong>of</strong> kidney tumors (adjusted from (Bjornsson et al. 1996; Dulaimi et al. 2004;<br />

Pavlovich and Schmidt 2004; Henske 2005))<br />

Histological<br />

type<br />

Presumed<br />

origin<br />

Clear cell RCC Proximal renal<br />

tubule<br />

Papillary RCC<br />

(basophilic<br />

and<br />

eosinophilic)<br />

Chromophobe<br />

RCC<br />

Proximal renal<br />

tubule<br />

Collecting duct<br />

(intercalated<br />

cells)<br />

Oncocytoma Collecting duct<br />

(intercalated<br />

cells)<br />

Collecting<br />

carcinoma<br />

Angiomyolipoma<br />

Frequent cytogenetic<br />

abnormalities in<br />

sporadic tumors *<br />

-3p, +5q, -Y, -8p, -9p, -<br />

14q, t(3;5)(p;q);<br />

+7, +17, -Y, +12, +16,<br />

+20<br />

-1, -2, -6, -10, -13, -17,<br />

-21<br />

-1, -Y, t(5;11)(q35;q13),<br />

t(9;11)(p23;q13)<br />

Collecting duct -1q32, -6p, -8p, -21q,<br />

highly aneuploid<br />

Mesenchymal<br />

cells (immature<br />

smooth muscle<br />

cells, fat cells)<br />

No specific<br />

chromosomal changes<br />

have been identified<br />

Frequent promoter<br />

hypermethylations in<br />

sporadic tumors<br />

VHL, RASSF1A,<br />

p16 INK4a , p14 ARF, APC,<br />

MGMT, GSTP1,<br />

RARß2, E-cadherin,<br />

TIMP3<br />

RASSF1A, p16 INK4a ,<br />

p14 ARF, APC, MGMT,<br />

GSTP1, RARß2, Ecadherin,<br />

TIMP3<br />

RASSF1A, p14 ARF,<br />

RARß2, TIMP3<br />

RASSF1A, p16 INK4a ,<br />

p14 ARF, APC, MGMT,<br />

GSTP1, TIMP3<br />

RASSF1A, p14 ARF,<br />

APC, MGMT, RARß2,<br />

TIMP3<br />

No epigenetic changes<br />

have been identified<br />

Hereditary genetic<br />

syndromes and<br />

implicated genes **<br />

Von Hippel-Lindau disease<br />

(VHL); Birt Hogg Dubé<br />

syndrome (BDH); Tuberous<br />

Sclerosis (TSC1, TSC2)<br />

Hereditary papillary renal<br />

carcinoma (c-MET);<br />

Hereditary leiomyomatosis<br />

renal cell cancer (FH);<br />

Hyperparathyroidism-jaw<br />

tumor (HRPT2); Tuberous<br />

Sclerosis (TSC1, TSC2)<br />

Birt Hogg Dubé syndrome<br />

(BDH); Tuberous Sclerosis<br />

(TSC1, TSC2)<br />

Birt Hogg Dubé syndrome<br />

(BDH); Tuberous Sclerosis<br />

(TSC1, TSC2)<br />

Hereditary leiomyomatosis<br />

renal cell cancer (FH)<br />

Tuberous Sclerosis (TSC1,<br />

TSC2)<br />

* Nomenclature cytogenetics: Loss <strong>of</strong> a chromosome (-); gain <strong>of</strong> a chromosome (+); translocation (t); short<br />

arm <strong>of</strong> a chromosome (p); long arm <strong>of</strong> a chromosome (q). ** Von-Hippel-Lindau (VHL); Birt Hogg Dubé<br />

(BDH), Fumarat hydratase (FH); Hyperparathroidism 2 (HRPT2); Tuberous Sclerosis 1 (TSC1), Tuberous<br />

Sclerosis 2 (TSC2)<br />

However, cytogenetic studies are mainly based on the analysis <strong>of</strong> gross tumors that may<br />

have gained numerous mutations after developing genetic instability. Thus, in most cases<br />

the initial genetic or epigenetic cause for the initiation <strong>of</strong> spontaneous kidney tumors<br />

remains elusive. However, studies <strong>of</strong> families with hereditary genetic syndromes have<br />

identified several predisposing genes involved in renal carcinogenesis. Due to the similar<br />

histology <strong>of</strong> hereditary and sporadic RCC, these genes were also suggested to be involved<br />

in the development <strong>of</strong> sporadic tumors (Table 1.1, right column).<br />

6


II: <strong>Kidney</strong> Tumors in Rats<br />

Chapter 1: Introduction<br />

Rats are the most common laboratory animal to study renal carcinogenesis. Although the<br />

incidence <strong>of</strong> spontaneous RCC in laboratory strains is very low (e.g. 0.08% in Sprague-<br />

Dawley and 0.28% in F344 rats) (Chandra et al. 1993), similar tumor types as those<br />

observed in humans have been experimentally induced in rodents by distinct chemicals,<br />

hormones, viruses and radiation (Hamilton 1975; Lock and Hard 2004). Studies in rodents<br />

not only allowed the analysis <strong>of</strong> the carcinogenic potential <strong>of</strong> numerous test compounds<br />

(see chapter 1.2.1) but also the identification <strong>of</strong> the earliest carcinogen-induced foci <strong>of</strong> cells<br />

(pre-neoplastic lesions) with alterations very similar to those found in end-stage tumors.<br />

From these findings it was concluded that kidney tumor development can occur in<br />

successive stages, from single atypical tubules to hyperplasia to adenoma and/or<br />

carcinoma (Figure 1.2) (Dietrich and Swenberg 1991). However, knowledge <strong>of</strong> the initial<br />

causes and the process <strong>of</strong> progression <strong>of</strong> chemically induced tumors is still limited. Current<br />

findings from rodent bioassays assume that not all carcinogen-induced pre-neoplastic<br />

lesions will progress to a neoplastic lesion and it is <strong>of</strong>ten not possible to distinguish lesions<br />

with a neoplastic potential from lesions that are self-limiting or even reversible, and thus<br />

may not have any significance for the testing <strong>of</strong> carcinogenicity (Alden and Kanerva 1982;<br />

Eustis 1989). To date, the potential contribution <strong>of</strong> pre-neoplastic lesions to carcinogeninduced<br />

tumor development is mainly assessed by histopathologic evaluation <strong>of</strong> similarities<br />

between compound-induced pre-neoplastic and neoplastic lesions, by the absence or<br />

presence <strong>of</strong> a morphological continuum between these lesions, and by the occurrence <strong>of</strong><br />

treatment-related cytotoxicity and regenerative cell proliferation (Eustis 1989).<br />

Common cytological alterations found in carcinogenic lesions in rats resemble those <strong>of</strong><br />

sporadic lesions in humans and are summarized in table 1.2. Both the chemical properties<br />

<strong>of</strong> the inducing compounds, as well as their metabolism and location <strong>of</strong> uptake within the<br />

different tubular segments, most likely determine the type and cellular origin <strong>of</strong> the lesions.<br />

For instance, clear cell/ acidophilic tumors in N-nitrosomorpholine (NNM)- treated rats were<br />

demonstrated to arise from the distal tubules and collecting ducts, which are also the main<br />

sites <strong>of</strong> NNM-induced acute cell damage (Bannasch et al. 1989; Nogueira et al. 1989).<br />

Similarly, the most common carcinogen-induced basophilic RCC as well as chromophobe<br />

RCC originate from the proximal tubules, which represent the most susceptible sites <strong>of</strong><br />

injury via metabolic activation <strong>of</strong> xenobiotica or their transporter-associated accumulation<br />

(e.g. ochratoxin A, fumonisine B1) (Boorman et al. 1992; Howard et al. 2001). Oncocytic<br />

cells were found to arise from the intercalated cells <strong>of</strong> the renal collecting duct <strong>of</strong> potassium<br />

7


Chapter 1: Introduction<br />

bicarbonate-, NNM-, streptomycin- or lead acetate-treated rats (Nogueira 1987; Mayer et al.<br />

1989; Lina and Kuijpers 2004).<br />

Preneoplastic and neoplastic lesions <strong>of</strong> mixed cellular phenotypes can occur spontaneously<br />

or subsequent to carcinogen treatment. However, it is not clear whether different cellular<br />

alterations occur by random hits in different tumor suppressor genes or oncogenes or by a<br />

transition process from one to another lesion type.<br />

Figure 1.2: Progression <strong>of</strong> renal lesions. Adjusted from Dietrich and Swenberg (Dietrich and<br />

Swenberg 1991). Microscopic images represent typical renal basophilic lesions <strong>of</strong><br />

Eker rats after exposure with renal carcinogens, and were obtained during the course<br />

<strong>of</strong> this thesis. For further information, see text.<br />

8


Chapter 1: Introduction<br />

Table 1.2: Representative images <strong>of</strong> frequent cellular abnormalities and biochemical features <strong>of</strong><br />

carcinogen-induced kidney tumors in rats. Adjusted from (Dietrich and Swenberg<br />

1991). Microscopic images kindly provided by Daniel R. Dietrich<br />

Histological type Morphological and cytological <strong>characteristics</strong> Representative image<br />

Clear cells Large translucent cytoplasm, irregular cell shape<br />

with small chromatin-dense nuclei. Cytoplasmatic<br />

accumulations <strong>of</strong> glycogen and lipids (Mayer et al.<br />

1988, Steinberg et al. 1992).<br />

Acidophilic<br />

(eosinophilic) cells<br />

Often associated with clear cells and are slightly<br />

larger than normal cells with a strongly eosinophilic<br />

cytoplasm<br />

Basophilic cells Basophilic cytoplasm. Increased abundance <strong>of</strong><br />

ribosomes, slightly enlarged nuclei with prominent<br />

nucleoli<br />

Chromophobic cells Enlarged cells with slightly translucent cytoplasm.<br />

Weak eosinophilic or basophilic staining. Extensive<br />

storage <strong>of</strong> mucopolysaccharides likely enclosed in<br />

cytoplasmatic vacuoles<br />

Oncocytic cells Large polygonal shape with acidophilic/<br />

eosinophilic and granular cytoplasm. Abundance<br />

<strong>of</strong> atypical mitochondria and a large content <strong>of</strong><br />

cytochrome c oxidase. Over-expression <strong>of</strong> GLUT1<br />

and hexokinase I.<br />

9


1.2 Renal Carcinogens<br />

Chapter 1: Introduction<br />

Based on the general assumption that multigenetic events are required for carcinogenesis,<br />

there are basically two possibilities by which a chemical carcinogen can increase the<br />

possibility <strong>of</strong> tumorigenesis within a tissue: 1) by increasing the rate <strong>of</strong> DNA or<br />

chromosomal damage, resulting in more errors per replication, or 2) by increasing the<br />

number <strong>of</strong> DNA replications and therefore the likelihood <strong>of</strong> spontaneous errors (Cohen and<br />

Arnold 2008). Initiated and clonally expanding cells might have different <strong>characteristics</strong> than<br />

their cell <strong>of</strong> origin and therefore might be differentially influenced by chemical carcinogens.<br />

For instance, it is not entirely clear whether progressing stages <strong>of</strong> transformed, neoplastic<br />

kidney cells are still capable <strong>of</strong> taking up or metabolizing carcinogens or rather<br />

dedifferentiate in favor <strong>of</strong> proliferation.<br />

Chemical carcinogens have been categorized according to two general mechanisms <strong>of</strong><br />

action into genotoxic and non-genotoxic carcinogens:<br />

1.2.1 Genotoxic Carcinogens<br />

Genotoxicity is induced by compounds that are able to directly interact with the DNA<br />

(mutagens), compounds that break chromosomes (clastogens) and compounds that<br />

damage the spindle apparatus (aneugens). Genotoxic carcinogens are either electrophilic<br />

molecules per se or are absorbed as stable pro-carcinogens that have to be bio-activated<br />

to their active electrophilic form, which can then covalently interact with any nucleophilic<br />

site in the cell, such as the DNA resulting in DNA adducts. Groups <strong>of</strong> genotoxins can<br />

therefore <strong>of</strong>ten be recognized by their structural features (e.g. alkenes, aromatic amines,<br />

nitrosamines or polycyclic aromatic hydrocarbons).<br />

According to their relatively simple mode <strong>of</strong> action, which involves a single chemical<br />

interaction with one specific target molecule, DNA adduct formation was expected as a<br />

linear function <strong>of</strong> the administered dose <strong>of</strong> the DNA-reactive compound. In fact most<br />

carcinogenicity studies that have used low doses <strong>of</strong> genotoxic compounds have been<br />

interpreted as showing no threshold (Lijinsky et al. 1988; Peto et al. 1991). It was therefore<br />

concluded that even the smallest doses <strong>of</strong> DNA-reactive compounds increase susceptibility<br />

to cancer and that a zero-risk or safe-dose may not be defined. However, it is now<br />

recognized that many factors e.g. efficiency <strong>of</strong> absorption, distribution, metabolism, DNA<br />

damage repair and mutation fixation, can affect the linear shape <strong>of</strong> the dose-response<br />

10


Chapter 1: Introduction<br />

curve, leading to a sublinear and supralinear curve progression (Williams et al. 1995;<br />

Swenberg et al. 2008). Due to these findings it is now frequently discussed, whether a<br />

threshold or no-observable-adverse-effect-level (NOAEL) may exist at least for some<br />

genotoxic compounds (Williams et al. 1995; Oesch et al. 2000; Hengstler et al. 2003).<br />

1.2.2 Non-Genotoxic Carcinogens<br />

Non-genotoxic carcinogens are a heterogeneous group <strong>of</strong> carcinogens that are not directly<br />

DNA-reactive. However, they are capable <strong>of</strong> inducing epigenetic effects on cells that 1)<br />

either indirectly result in DNA modification and subsequently pro-carcinogenic mutations, or<br />

2) promote the development <strong>of</strong> initiated cells into neoplasms. Numerous tumor promoting<br />

mechanisms have been identified for a wide range <strong>of</strong> non-genotoxic compounds. Many <strong>of</strong><br />

these are cell type-, tissue-, sex- or species-dependent and usually require sustained and<br />

high (above threshold) exposures to induce a carcinogenic effect. For kidney<br />

carcinogenesis non-genotoxic carcinogens can be categorized into three basic groups:<br />

I: Cytotoxic Carcinogens<br />

A number <strong>of</strong> non-genotoxic carcinogens were demonstrated to induce tumors at high<br />

cytotoxic doses. It is assumed that exposure to cytotoxic carcinogens results in tissue<br />

damage, and the subsequent regenerative cell proliferation may either contribute to the<br />

fixation <strong>of</strong> spontaneous mutations, or to the promotion <strong>of</strong> naturally occurring tumors in<br />

tissues with a high spontaneous tumor rate (Dietrich and Swenberg 1990; Cohen and<br />

Arnold 2008). One <strong>of</strong> the best studied species-, sex- and tissue-specific mechanisms is the<br />

chemically induced α2u-globulin nephropathy, which is associated with the onset <strong>of</strong> renal<br />

neoplasms in male rats (Swenberg 1993). α2u-globulin is a plasma protein that binds<br />

hydrophobic small molecules for subsequent absorption and lysosomal degradation by the<br />

P2 segment <strong>of</strong> the renal proximal tubule. Binding <strong>of</strong> xenobiotica, e.g. unleaded petrol,<br />

decalin or d-limonene, may result in a structural alteration <strong>of</strong> α2u-globulin and its protection<br />

from lysosomal degradation. Subsequent accumulation <strong>of</strong> lysosomal protein droplets in the<br />

kidney frequently results in tubular necrosis, compensatory cell proliferation (regeneration),<br />

and an increased chance for the fixation <strong>of</strong> spontaneous genetic damage and therefore the<br />

onset <strong>of</strong> renal tumorigenesis (Dietrich and Swenberg 1990; Swenberg and Lehman-<br />

McKeeman 1999; Dill et al. 2003).<br />

11


II: Non-Cytotoxic Carcinogens<br />

Chapter 1: Introduction<br />

Non-cytotoxic carcinogens exert their tumor promoting potential without causing prior cell<br />

loss. Instead, they are able to influence a variety <strong>of</strong> cellular signaling pathways that are<br />

<strong>of</strong>ten controlled by tumor suppressor genes and proto-oncogenes. Non-cytotoxic<br />

carcinogens may cause a cell to commence cell division by triggering mitosis or inhibiting<br />

apoptosis. Such ‘mitogens’ <strong>of</strong>ten mimic the activation <strong>of</strong> extracellular growth factors by<br />

either directly activating the corresponding receptor, or by modulating downstream<br />

signaling events through interaction with kinases, phosphatases, scaffold proteins or<br />

chaperons, ultimately inducing mitosis (Perona 2006; Lawrence et al. 2008). Other noncytotoxic<br />

carcinogens can inhibit intercellular communication via gap junctions and affect<br />

cellular growth regulation and differentiation (Chipman et al. 2003; Leithe et al. 2006).<br />

III: Indirect genotoxic carcinogens<br />

The induction <strong>of</strong> oxidative stress has been proposed as one possible mechanism <strong>of</strong> action<br />

for non-genotoxic carcinogens, either by increasing the production <strong>of</strong> reactive oxygen<br />

species (ROS), or by inhibiting the antioxidant capability <strong>of</strong> the target cell. ROS can not only<br />

modify cellular lipids and proteins but also directly oxidize and damage the DNA. The best<br />

studied oxidative DNA lesion is 8-hydroxydeoxyguanosine (OH8dG) which occurs from<br />

oxidation <strong>of</strong> guanine at the C8 position within the DNA, or from oxidation <strong>of</strong> dGTP and its<br />

later incorporation into the DNA. Both can result in site-specific mutagenesis (Shibutani et<br />

al. 1991; Hussain and Harris 1998). Recent evidence shows that ROS and oxidative DNA<br />

damage can modulate DNA methylation patterns (Turk et al. 1995) which may result in<br />

epigenetic silencing <strong>of</strong> tumor suppressor genes or activation <strong>of</strong> oncogenes. Besides their<br />

role in tumor initiation, ROS were further shown to be involved in tumor promotion and<br />

progression. High doses <strong>of</strong> cellular ROS or ROS-promoting agents are clearly cytotoxic and<br />

can result in regenerative cell proliferation and fixation <strong>of</strong> mutations. Low doses <strong>of</strong> ROS<br />

may contribute to abnormal gene expression, cell signaling and alteration <strong>of</strong> second<br />

messenger systems. For instance, ROS can activate both mitogen-activated protein (MAP)<br />

kinase/AP-1 and NFkappa-B pathways, which are critically involved in cell proliferation and<br />

apoptosis (Klaunig and Kamendulis 2004).<br />

12


1.3 Renal Carcinogens Relevant for this Study<br />

Chapter 1: Introduction<br />

For this thesis, three naturally occurring renal carcinogens were used and the following<br />

chapter will give an overview <strong>of</strong> the chemical and biological properties <strong>of</strong> the respective<br />

compounds.<br />

1.3.1 Aristolochic acid<br />

Aristolochic acid (AA) represents a mixture <strong>of</strong> aristolochic acid I (AAI) and aristolochic acid<br />

II (AAII) (Figure 1.3) and is primarily found in the Aristolochia and Asarum species <strong>of</strong> the<br />

Aristolochiaceae family <strong>of</strong> plants. It can be found in a number <strong>of</strong> botanical products such as<br />

traditional medicines, dietary supplements and weight loss remedies.<br />

Figure 1.3: Chemical structures <strong>of</strong> aristolochic acid I and aristolochic acid II, from<br />

(Balachandran et al. 2005)<br />

I: Acute and Chronic Toxicity<br />

The kidneys are the primary target organ for acute and chronic AA toxicity in both<br />

laboratory animals (rats, mice and rabbits) and humans. In rats, the oral LD50 (the quantity<br />

<strong>of</strong> a compound per kg per body weight (bw) that kills 50% <strong>of</strong> the study population) was<br />

reported to be 203.4 mg/kg bw in males and 183.9 mg/kg bw in females (Mengs 1987).<br />

Rats treated with a single or continuous high oral doses 1-10mg/kg BW) <strong>of</strong> AA developed<br />

renal failure with tubular necrosis <strong>of</strong> the corticomedullary junction (Mengs 1987; Mengs and<br />

Stotzem 1993), and had elevated plasma creatinine and urea levels (Mengs and Stotzem<br />

1993; Liu et al. 2003; Cui et al. 2005). In addition, in some studies interstitial fibrosis was<br />

observed (Debelle et al. 2002; Sun et al. 2006), although this finding could not be replicated<br />

13


Chapter 1: Introduction<br />

by others (Cosyns et al. 1998; Cui et al. 2005). In humans, consumption <strong>of</strong> AA-containing<br />

botanical products has been associated with the onset <strong>of</strong> aristolochic acid nephropathy,<br />

characterized by mild tubular proteinuria, extensive interstitial fibrosis, tubular atrophy,<br />

global sclerosis <strong>of</strong> glomeruli, and rapid progression to renal failure (Nortier and<br />

Vanherweghem 2002; Cosyns 2003).<br />

II: Carcinogenicity<br />

AA is mutagenic and carcinogenic in mice, rats, rabbits and humans (Arlt et al. 2002).<br />

Recently, AA-containing products were classified as carcinogenic to humans (Group 1) by<br />

the International Agency for Research on <strong>Cancer</strong> (IARC) (IARC 2002b). In rats, application<br />

<strong>of</strong> high oral doses (50mg/kg bw) for three days resulted in increased incidences <strong>of</strong><br />

preneoplastic and neoplastic lesions <strong>of</strong> the kidney after a latency <strong>of</strong> 6 months (Cui et al.<br />

2005). In another study, continuous gavage with 1-10mg/kg bw for 3 to 6 months resulted<br />

in a dose-dependent increase <strong>of</strong> a variety <strong>of</strong> tumors predominantly observed in the<br />

forestomach, the kidneys (epithelial adenomas and adenocarcinomas <strong>of</strong> the renal cortex)<br />

and tumors <strong>of</strong> the renal pelvis or urinary bladder (Mengs et al. 1982; Cosyns et al. 1999)<br />

A number <strong>of</strong> single case reports as well as studies from a large cohort <strong>of</strong> patients <strong>of</strong> a<br />

Belgian slimming regimen, who received AA-contaminated products by accident, have<br />

reported a high prevalence <strong>of</strong> urothelial cancer in patients with aristolochic acid<br />

nephropathy (AAN) (Cosyns et al. 1999; Nortier et al. 2000). Most recently, AA was also<br />

associated with the etiology <strong>of</strong> the Balkan Endemic Nephropathy (BEN), a form <strong>of</strong> interstitial<br />

fibrosis associated with urothelial tumors which is most prevalent in the Balkan countries<br />

Bosnia, Serbia Croatia, Romania and Bulgaria (Arlt et al. 2007; Grollman et al. 2007).<br />

However, until now it has not been unequivocally established that the inhabitants <strong>of</strong> BEN<br />

areas are indeed exposed to higher concentration <strong>of</strong> AA than inhabitants <strong>of</strong> other regions,<br />

and the routes <strong>of</strong> exposure remain largely unknown.<br />

The genotoxic action <strong>of</strong> AA is based on its bioactivation to an intermediate aristolactamnitrium<br />

ion (Krumbiegel et al. 1987; Chan et al. 2006; Stiborova et al. 2008), which is able<br />

to form bulky adducts at purine bases <strong>of</strong> the DNA (Schmeiser et al. 1988), including<br />

dAdenin-AAI adducts which were identified as the main effectors for the mutagenic and<br />

carcinogenic action <strong>of</strong> AA (Pfau et al. 1990; Pfau et al. 1991). The mutagenic potential for<br />

AA was demonstrated in a number <strong>of</strong> in vitro systems (Schmeiser et al. 1984; Arlt et al.<br />

2002; Zhang et al. 2004). Increased mutation frequencies were also seen in the kidneys,<br />

bladder and forestomach <strong>of</strong> carcinogenicity models such as the transgenic Muta TM Mouse<br />

(Kohara et al. 2002) and Big Blue rats (Mei et al. 2006). In another study, analysis <strong>of</strong><br />

14


Chapter 1: Introduction<br />

mutational spectra from tumors <strong>of</strong> AA-treated rats revealed activating AT to TA transversion<br />

mutations in codon 61 <strong>of</strong> the H-ras oncogene in forestomach, lung and ear duct tumors, but<br />

not in kidney tumors (Schmeiser et al. 1990), suggesting an H-ras independent pathway <strong>of</strong><br />

renal tumor induction. In humans, AT to TA transversion mutations were observed in the<br />

p53 tumor suppressor gene in urothelial tumors derived from AAN and BEN patients (Lord<br />

et al. 2004; Arlt et al. 2007).<br />

1.3.2 Ochratoxin A<br />

Ochratoxin A (OTA) is a naturally occurring mycotoxin, commonly produced by a variety <strong>of</strong><br />

Penicillium and Aspergillus species (Figure 1.4). It occurs as a notorious contaminant <strong>of</strong><br />

grains and cereals and is also commonly found in food and beverages, e.g. in dried fruits,<br />

chocolate, juices, wine or beer (Kuiper-Goodman 1991; Clark and Snedeker 2006).<br />

Figure 1.4: Chemical structure <strong>of</strong> ochratoxin A. From: www.wikipedia.org/wiki/Ochratoxin<br />

I: Acute and Chronic Toxicity<br />

Acute toxicity <strong>of</strong> OTA is highly species-specific with an oral LD50 ranging from 0.2-<br />

1.0mg/kg bw for species such as sheep, pigs, rabbits and dogs, to 20mg/kg bw in rats, and<br />

to 46-58mg/kg bw in mice. In rats, chronic exposure with OTA targets primarily the kidney.<br />

OTA accumulates to high amounts in the proximal tubule where it provokes morphological<br />

changes predominantly in the S3 segment <strong>of</strong> the proximal convoluted tubule, including<br />

cytoplasmatic alterations, karyomegaly, and tubular degeneration as well as the<br />

development <strong>of</strong> cortical fibrosis. Functional alterations in response to OTA treatment are<br />

reflected by a decreased creatinine, inulin and p-aminohippurate clearance, polyuria,<br />

glucosuria, and a decreased osmolarity <strong>of</strong> the urine (NTP 1989).<br />

15


II: Carcinogenicity<br />

Chapter 1: Introduction<br />

OTA is a potent renal carcinogen in rodents (NTP 1989; Boorman et al. 1992; Rasonyi et<br />

al. 1999; Mantle et al. 2005). However, due to inadequate evidence for carcinogenicity in<br />

humans, it was classified by IARC as possibly carcinogenic to humans (group 2B). Rats<br />

exposed for two years with 210µg/kg bw/day developed a massive increase <strong>of</strong> benign and<br />

malignant tumors arising from tubular renal epithelial cells (Boorman et al. 1992). The 60%<br />

tumor incidence in male rats was 10-fold higher than observed in female rats, suggesting a<br />

sex-specific mode <strong>of</strong> action. To date, the mechanism <strong>of</strong> OTA-induced carcinogenicity is still<br />

not well defined and controversial results regarding a genotoxic (Faucet et al. 2004) or a<br />

non-genotoxic (Mally et al. 2005) mode <strong>of</strong> action have been published. Since OTA has not<br />

been clearly demonstrated to form reactive intermediates capable <strong>of</strong> covalently interacting<br />

with the DNA (Kamp et al. 2005; Mally et al. 2005; Delatour et al. 2008), a non-genotoxic<br />

mechanism <strong>of</strong> action is suggested by the majority <strong>of</strong> studies. Recent data indicate that renal<br />

toxicity as well as the observed DNA damage are most likely attributable to oxidative stress<br />

(Kamp et al. 2005; Marin-Kuan et al. 2006; Domijan et al. 2007). Notably, MAPK-ERK<br />

pathway activation as well as histone deacetylase-induced gene silencing are suggested to<br />

play an important role in OTA-induced tumorigenesis (Marin-Kuan et al. 2007).<br />

Similar to aristolochic acid, OTA is assumed to be involved in the onset <strong>of</strong> BEN, based on<br />

studies that revealed a higher frequency <strong>of</strong> BEN in inhabitants <strong>of</strong> areas endemic for OTAcontaining<br />

foods (Fuchs and Peraica 2005; Clark and Snedeker 2006). Moreover, OTA was<br />

more frequently detected in blood samples <strong>of</strong> patients with BEN than in healthy individuals<br />

(Fuchs and Peraica 2005). Based on data from one human volunteer who ingested 395ng<br />

<strong>of</strong> radioactivelly labelled [3H]-OTA, the half-life <strong>of</strong> 35,5 days was about eight-fold longer the<br />

OTA half-life determined for rats (Studer-Rohr et al. 2000). Therefore, although a causal<br />

relationship between OTA and the onset <strong>of</strong> BEN and urothelial tumors still remains elusive,<br />

the long half-life in humans might contribute to the accumulation <strong>of</strong> OTA in various target<br />

organs, and thus to the onset <strong>of</strong> BEN or OTA-mediated cancers.<br />

1.3.3 Cycasin / Methylazoymethanol<br />

Methylazoxymethanol (MAM) is the biologically active metabolite <strong>of</strong> cycasin (Figure 1.5, A),<br />

which is a toxic glycoside <strong>of</strong> the cycad plants Cycas circinalis and Cycas revoluta. Nuts,<br />

roots and leaves <strong>of</strong> these plants have been sources for food and medicine for the<br />

indigenous people <strong>of</strong> the Western pacific (e.g. Guam).<br />

16


A: B: C:<br />

Chapter 1: Introduction<br />

Figure 1.5: Chemical structures <strong>of</strong> (A) cycasin, (B) MAM and (C) methyldiazonium ion. From:<br />

(Sohn et al. 2001)<br />

I: Acute and Chronic toxicity<br />

The acute oral LD50 <strong>of</strong> cycasin is highly species-dependent, ranging from less than 20<br />

mg/kg bw in the guinea pig to 562 mg/kg bw in the rat (Hirono 1972). In rats, a 6.5 fold<br />

lower oral LD50 was detected when the active metabolite MAM was directly gavaged<br />

(Hirono 1972). Acute toxic effects <strong>of</strong> MAM and cycasin mainly occurred in the liver,<br />

including glycogen, RNA and phospholipid depletion, cellular necrosis, and haemorrhage<br />

(Williams and Laqueur 1965). Moreover, MAM was described as a developmental<br />

neurotoxin in rats when administered during fetal or neonatal periods <strong>of</strong> CNS development<br />

(Singh 1980). In humans, consumption <strong>of</strong> cycasin-containing products resulted in liver<br />

damage and was associated with the etiology <strong>of</strong> the neurological disorder Amyotrophic<br />

Lateral Sclerosis-Parkinson Dementia Complex (ALS-PDC) in people <strong>of</strong> the Western<br />

Pacific (Ince and Codd 2005; Borenstein et al. 2007).<br />

II: Carcinogenicity<br />

Cycasin was found to be a potent genotoxic carcinogen in rodents, and the duration <strong>of</strong><br />

exposure was shown to highly influence the site <strong>of</strong> tumor development (Hirono 1972). In<br />

cycasin treated rats, the target sites <strong>of</strong> tumor development w highly dependent on the dose<br />

regimen. In rats, one single high oral doses <strong>of</strong> cycasin (100 to 500 mg/kg bw) (Hirono et al.<br />

1968; Laqueur 1968; Fukunishi et al. 1971; Uchida and Hirono 1981) or repeated oral<br />

doses (25mg/kg bw for 12 weeks) (Fukunishi et al. 1971) predominantly resulted in tumors<br />

<strong>of</strong> the kidneys, followed by neoplasms <strong>of</strong> the liver and colon. In contrast, life long exposure<br />

to lower doses <strong>of</strong> cycasin (4mg/kg bw via drinking water) primarily induced mammary and<br />

testicular cancer, while tumors <strong>of</strong> the liver, kidney and intestine only occurred sporadically<br />

(Fukunishi et al. 1971). Cycasin was carcinogenic only after passage through the<br />

gastrointestinal tract (Laqueur 1964), where it was deglucosylated by ß-glucosidases <strong>of</strong> the<br />

17


Chapter 1: Introduction<br />

gut micr<strong>of</strong>lora to release the principal metabolite MAM (Spatz et al. 1967; Matsushima et al.<br />

1979; Choi et al. 1996). Under physiological conditions, MAM spontaneously hydrolyzes to<br />

water, formaldehyde, and the “ultimate carcinogen” methyldiazonium ion (Figure 1.5, C), an<br />

active DNA methylating agent, responsible for N7-methylguanine- and O6-methylguanine-<br />

DNA-alkylations, as observed in vitro (Matsumoto and Higa 1966) and in vivo (Zedeck and<br />

Brown 1977; Sohn et al. 2001) after MAM exposure. However, to date a reliable<br />

determination <strong>of</strong> the respective adducts in kidneys <strong>of</strong> MAM-treated rats remains difficult.<br />

Intragastric or intraperitoneal administration <strong>of</strong> MAM induced similar tumors as observed<br />

with cycasin, although with a 15% higher incidence (Laqueur 1968). Due to the chemical<br />

instability <strong>of</strong> MAM most studies were performed using synthetic MAM acetate (MAMAc).<br />

MAMAc is highly sensitive to hydrolysis by various blood and organ esterases, and<br />

therefore is deacetylated to MAM almost immediately following administration (Zedeck and<br />

Brown 1977).<br />

Up to now, no case reports <strong>of</strong> cancer in humans exposed to cycasin or MAM have been<br />

reported. Since further epidemiological studies did not show increased cancer mortality 2 to<br />

7 years after heavy intake <strong>of</strong> cycad plants or plant products, cycasin and MAM were<br />

classified by the IARC as possibly carcinogenic to humans (group 2B carcinogen).<br />

1.4 Methods to Identify Renal Carcinogens<br />

The vast majority <strong>of</strong> all kidney cancers are expected to be caused by exogenic factors, like<br />

exposure to industrial or environmental chemicals, pesticides or mycotoxins (McLaughlin<br />

and Lipworth 2000). Identification <strong>of</strong> potential carcinogens and assessing their risk for<br />

humans is therefore the first step in disease prevention. For decades, methods for the<br />

identification <strong>of</strong> renal and other carcinogens have been extensively developed, including<br />

the rodent bioassays (see below), animal models for cancer induction at distinct organ sites<br />

(e.g. the Eker rat model <strong>of</strong> renal cancer, for details see chapter 1.4.3), in vitro methods to<br />

study the effects <strong>of</strong> carcinogens in cells and organ slice cultures, and mechanistic assays to<br />

study various molecular events such as metabolic activation and detoxification <strong>of</strong><br />

carcinogens, DNA damage and repair and mutagenicity.<br />

18


1.4.1 Standard carcinogenicity testing<br />

Chapter 1: Introduction<br />

For almost 40 years, carcinogenicity testing has been used in an attempt to predict the<br />

potential <strong>of</strong> chemicals and pharmaceuticals to cause cancer in humans. According to<br />

different guidelines in the United States, Japan and Europe, carcinogenicity testing is<br />

required when human exposure to the respective compound is expected in a continuing or<br />

intermittent manner for at least three months (according to the U.S. Food and Drug<br />

Administration (FDA)) or six months (according to Japan’s Guidelines for Toxicity Studies <strong>of</strong><br />

Drugs Manual, or to European rules governing medicinal products, respectively). At any<br />

rate, carcinogenicity testing is required when there is concern about the potential <strong>of</strong> a<br />

chemical known to have a carcinogenic effect (based e.g. on structure activity<br />

relationships).<br />

The standard carcinogenicity testing procedure consists <strong>of</strong> a battery <strong>of</strong> genotoxicity tests<br />

involving: 1) a bacterial reverse mutation test (Ames test), 2) an in vitro genotoxicity test<br />

(e.g. in vitro mammalian chromosome aberration test, in vitro mammalian cell gene<br />

mutation test, or unscheduled DNA synthesis test in mammalian cells), and 3) an in vivo<br />

genotoxicity test (e.g. mammalian erythrocyte micronucleus test, mammalian bone marrow<br />

chromosome aberration test or unscheduled DNA synthesis test with mammalian liver cells<br />

in vivo).<br />

To be designated as a genotoxic carcinogen, compounds must have been proven positive<br />

in the Ames test combined with at least one in vitro and one in vivo genotoxicity test. Only<br />

compounds with negative or substantially negative results in these tests are further<br />

evaluated in the rodent lifetime bioassay for their carcinogenic potential.<br />

1.4.2 The lifetime rodent bioassay<br />

To date almost 1500 chemicals have been tested by a standard protocol originally<br />

developed by the National Toxicology Program (NTP). The protocol involves exposing<br />

distinct strains <strong>of</strong> rats (Sprague Dawley or Wistar rats) and mice (B6C3F1) <strong>of</strong> both sexes to<br />

the respective compound for a period <strong>of</strong> 104 weeks. Typically 50 animals per sex and dose<br />

group and 4 doses, i.e. 0 (control), the maximum tolerated dose (MTD), MTD/2 and MTD/4<br />

are used The MTD <strong>of</strong> a test compound is generally determined from a preceding 90-day<br />

study using the route and method <strong>of</strong> administration that will be used in the lifetime<br />

bioassay. During the two year bioassay, animals <strong>of</strong> all groups are dosed for the indicated<br />

19


Chapter 1: Introduction<br />

time and subsequently sacrificed for a post-mortem evaluation <strong>of</strong> tumor incidences and<br />

pathological changes in 24 target organs (OECD 1981; NTP 1992).<br />

Utilizing the rodent bioassay for identifying potential cancer hazards for humans is generally<br />

based on two assumptions: a) that high dose effects can be extrapolated to lower doses<br />

and b) that an interspecies extrapolation between animals and humans is possible (Ward<br />

2007). The latter assumption may be reasonable at least for genotoxic compounds that<br />

tend to be carcinogenic across species, in both sexes and at all dose levels (e.g. aflatoxin<br />

B1 or vinyl chloride). In contrast, non-genotoxic carcinogens <strong>of</strong>ten do not induce adverse<br />

effects if dosed below a certain threshold (see 1.2.2) and may act via species-specific<br />

mechanisms that have no relevance for humans. Since current standard test procedures<br />

are based on high-dose regimens to warrant a potential detection <strong>of</strong> carcinogenic<br />

properties <strong>of</strong> a compound, an overestimation <strong>of</strong> the carcinogenic potential <strong>of</strong> a testsubstance<br />

is possible.<br />

The long duration <strong>of</strong> the rodent bioassay together with the high number <strong>of</strong> animals leads to<br />

enormous costs. Especially for drug development, low specificity <strong>of</strong> the standard test<br />

battery is a major drawback, and promising drug candidates may be taken out at late<br />

stages <strong>of</strong> the developmental process when tested “false positive”. In case <strong>of</strong> xenobiotica,<br />

positive carcinogenicity testing determines the intensity <strong>of</strong> regulatory management and<br />

restriction <strong>of</strong> marketing.<br />

To improve in vivo carcinogenicity testing without compromising human safety, current<br />

efforts try to augment the depth <strong>of</strong> studies by introducing more mechanistic data. Moreover,<br />

by adopting the directive on the protection <strong>of</strong> experimental animals in 1986, the protection<br />

and welfare <strong>of</strong> animals has become a European value. The European Union and its<br />

Member States are obliged to ensure that the number <strong>of</strong> experimental animals, their<br />

possible pain, suffering, distress or lasting harm as a consequence <strong>of</strong> procedures being<br />

conducted upon them, shall be kept to an absolute minimum. The directives thus can be<br />

summarized as the “3R- principle”, to reduce, replace and refine animal experiments.<br />

Recently, international governmental organizations and industry partners agreed upon the<br />

new International Conference on Harmonization (ICH) guideline “Testing for<br />

Carcinogenicity <strong>of</strong> Pharmaceuticals” (ICH 1997). This new guidance allows the use <strong>of</strong> only<br />

one rodent species (preferably the rat) and the replacement <strong>of</strong> the second species by newly<br />

developed models like the newborn mouse assay or one <strong>of</strong> various transgenic mouse<br />

models (e.g. the p53+/- mouse). The alternative usage <strong>of</strong> genetically engineered models<br />

might also provide additional mechanistic information not readily available from the<br />

20


Chapter 1: Introduction<br />

conventional long-term assay. In addition, the ‘International Agency for Research on<br />

<strong>Cancer</strong>’ working group has recommended the use <strong>of</strong> preneoplastic lesions as early<br />

indicators for carcinogenic activity (Williams 1999).<br />

Other promising model systems under development include non-mammalian species, in<br />

vitro cell systems, computer-based predictive toxicology models, or microarray-based<br />

toxicogenomics (described in chapter 1.6.). However, to allow reliable dose- and speciesextrapolation<br />

through the use <strong>of</strong> these systems requires a much more in-depth<br />

understanding <strong>of</strong> mechanistic processes involved in the onset and progression <strong>of</strong><br />

chemically induced carcinogenesis<br />

1.4.3 The Eker Rat Model <strong>of</strong> Renal Carcinogenesis<br />

Eker rats are a rodent model <strong>of</strong> hereditary kidney cancer. Their hereditary tumors resemble<br />

chemically induced tumors in other rat strains, as well as human renal adenomas and<br />

carcinomas. Thus, Eker rats were applied to study the etiology and mechanisms underlying<br />

kidney cancer. Their genetic predisposition is caused by a heterozygous insertion <strong>of</strong> a<br />

retroviral element into the tuberous sclerosis 2 (TSC2) tumor suppressor gene (Yeung et al.<br />

1994; Hino et al. 1995). Under physiological conditions, TSC2 encodes a protein called<br />

tuberin that binds to the TSC1 gene product hamartin to form a complex which acts as a<br />

GTPase-activating protein (GAP), thereby preventing Rheb-GTP-dependent stimulation <strong>of</strong><br />

the PI3K-Akt-Tsc1/2-Rheb-mTor pathway. Consequently, TSC2 and TSC1 are key<br />

mediators <strong>of</strong> growth factors or various nutrients to regulate cell growth, proliferation,<br />

migration and differentiation (Mak and Yeung 2004; Jozwiak 2006; Wullschleger et al.<br />

2006; Hanna et al. 2008).<br />

The inheritance <strong>of</strong> the TSC2 mutation follows Mendelian genetics. While a homozygous<br />

loss <strong>of</strong> the TSC2 gene causes embryonic lethality, the heterozygous <strong>of</strong>fspring is<br />

predisposed to the spontaneous development <strong>of</strong> multiple and <strong>of</strong>ten bilateral RCC with a<br />

100% incidence by one year <strong>of</strong> age (Everitt et al. 1995). Loss <strong>of</strong> heterozygosity (LOH) was<br />

only seen in 60% <strong>of</strong> the renal tumors in Eker rats, suggesting that a second genetic hit<br />

within the TSC2 gene is not the exclusive rate-limiting step for the development <strong>of</strong> renal cell<br />

carcinomas in this strain (Yeung, 1995). Rather, alternative pathogenic mechanisms such<br />

as haploinsufficiency seem possible.<br />

Additional studies have demonstrated that Eker rats are highly susceptible towards<br />

genotoxic and non-genotoxic renal carcinogens, which makes them a useful model for the<br />

21


Chapter 1: Introduction<br />

early detection <strong>of</strong> renal carcinogens. Eker rats treated with a single dose <strong>of</strong> the DNA<br />

alkylating dimethylnitrosamine (DMN) had a 70-fold higher average number <strong>of</strong> tumors per<br />

animal, compared to DMN-treated wild type rats (Walker, 1992). Furthermore,<br />

transplacental administration <strong>of</strong> N-ethyl-nitrosamine (ENU) resulted in the early occurrence<br />

<strong>of</strong> gross RCC after 8 weeks <strong>of</strong> age (Hino et al. 1993). No increased incidence but an<br />

advanced progression <strong>of</strong> preneoplastic lesion was observed in Eker rats that were<br />

subchronically treated with the tumor promoter sodium barbital (Wolf et al. 1998). In<br />

contrast, genotoxic non-renal carcinogens had no consistent effect on total numbers or<br />

incidence <strong>of</strong> preneoplastic and neoplastic lesions in the Eker rat kidney (Morton et al.<br />

2002). From these studies, it was concluded that Eker rats represent a useful tool to identify<br />

renal carcinogens and to study the underlying mechanisms.<br />

1.5 Toxicogenomics<br />

Microarrays have been used as a powerful and widespread tool for the simultaneous<br />

measurement <strong>of</strong> gene expression from thousands <strong>of</strong> genes within a target tissue. Soon<br />

after the first microarrays were applied to analyze molecular events that underlie chemically<br />

induced toxicity (toxicogenomics), toxicologists became fascinated by the multitude <strong>of</strong><br />

possibilities for safety assessment: Gene expression data may be used to identify<br />

mechanistic pathways involved in the dose-, tissue-, inter- and intra-specific heterogeneity<br />

<strong>of</strong> compound-induced toxicity. In addition, toxicogenomics can be used to identify and<br />

characterize novel biomarkers which might serve as early predictors for adverse effects <strong>of</strong><br />

test compounds in short term in vivo or in vitro experiments (Luhe et al. 2005).<br />

1.5.1 Principles <strong>of</strong> the Microarray Technology<br />

To date various types <strong>of</strong> microarrays are commercially available, such as cDNA arrays or<br />

oligonucleotide arrays. Currently Affymetrix GeneChips® are the most popular commercial<br />

arrays, and were also used for this thesis.<br />

Affymetrix GeneChips® consist <strong>of</strong> up to 1.3 million 25-mer oligonucleotide probes attached<br />

to a quartz surface, each with a gene-specific DNA sequence able to bind complementary<br />

target RNA <strong>of</strong> a test sample. Probe-target hybridizations can be quantified to determine the<br />

22


Chapter 1: Introduction<br />

relative abundance <strong>of</strong> an expressed gene in a test sample. The main feature <strong>of</strong> Affymetrix<br />

chips is that each gene is represented by an ensemble <strong>of</strong> probes (probe set) mapping to<br />

eleven different regions <strong>of</strong> the gene (Figure 1.6, A). Each <strong>of</strong> the eleven oligonucleotides is<br />

furthermore represented by a set <strong>of</strong> perfect match probes (PM) and identical mismatched<br />

probes (MM), with the exception <strong>of</strong> a single base difference in a central position. The<br />

mismatch probes serve as controls to determine the specificity <strong>of</strong> target-probe<br />

hybridizations and therefore to receive a higher reliability <strong>of</strong> gene expression data. As an<br />

example, Affymerix Rat Genome RAE_230_2 GeneChips® used for this thesis cover the<br />

entire genome <strong>of</strong> the rat by containing 30.199 probe sets encoding known annotated genes<br />

and unknown open reading frames (expressed sequence tags (EST)).<br />

Figure 1.6: (A) Composition <strong>of</strong> an Affymetrix chip. MM: mismatch oligonucleotide; PM: perfect<br />

match oligonucleotide. (B) Overview on Affymetrix chip hybridization. (C) Staining <strong>of</strong><br />

the DNA-cRNA hybrides using streptavidin-phycoerythrin. Modified from Affymetrix<br />

Expression Analysis: Technical manual for eukaryotic sample and array processing.<br />

23


Chapter 1: Introduction<br />

For gene expression analysis using Affymetrix chips, RNA is isolated from different test<br />

samples for subsequent cDNA preparation using a T7-Oligo(dT) promoter primer (Figure<br />

1.6, B). Following RNase H-mediated second-strand cDNA synthesis, the double-stranded<br />

cDNA serves as a template for in vitro transcription (IVT). The IVT reaction is carried out in<br />

the presence <strong>of</strong> T7 RNA polymerase and a mix <strong>of</strong> biotinylated nucleotides for<br />

complementary RNA (cRNA) amplification and biotin labeling. The biotinylated cRNA<br />

targets are then cleaned up, fragmented, and hybridized onto an Affymetrix GeneChip<br />

expression array (Figure 1.6, B). Biotinylated probe-target hybridizations are visualized by<br />

staining the arrays with a streptavidin/phycoerythrin solution and the addition <strong>of</strong> a<br />

biotinylated anti-streptavidin antibody, followed by a second streptavidin/phycoerythrin<br />

staining step (Figure 1.6, C). Hybridization and staining <strong>of</strong> Affymetrix arrays are performed<br />

with specialized robots, which increase both reproducibility and throughput.<br />

Fluorescence signals can be scanned and raw data image files (DAT) can be converted<br />

into CEL-files using Affymetrix Microarray Suite (MAS) 5.0 algorithm. In CEL files, the scan<br />

data from 36 pixels per PM/ MM probe set are averaged, background, gradients and<br />

distortions are corrected. The fluorescent intensities <strong>of</strong> the eleven PM/MM probe pairs per<br />

gene are then condensed to one intensity value per probe set, associated with a statistical<br />

detection <strong>of</strong> a p-value calculated from the intensity differences <strong>of</strong> the PM and corresponding<br />

MM oligonucleotides. This p-value indicates how reliably a transcript is detected and<br />

transcripts below a distinct quality setting p-value can be excluded from further statistical<br />

analysis. Different statistical tests can then be used to elucidate significant gene<br />

deregulations between two or more test samples.<br />

1.5.2 Microarrays- a Novel Tool to Study Carcinogenesis<br />

I: Diagnostic approach<br />

Microarrays have been used to study renal carcinogenesis and a number <strong>of</strong> publications<br />

have focused on the molecular classification <strong>of</strong> the different subtypes <strong>of</strong> kidney cancers in<br />

humans (Takahashi et al. 2006; Furge et al. 2007; Young et al. 2008). These studies not<br />

only revealed specific gene expression pr<strong>of</strong>iles for different histopathological subtypes, but<br />

also <strong>of</strong>fered a more detailed insight into the underlying pathways. In addition, a number <strong>of</strong><br />

studies have suggested the use <strong>of</strong> microarrays as promising tool for refining the diagnosis<br />

and staging <strong>of</strong> RCC, as well as for highlighting potential therapeutic targets (Tan et al.<br />

2004).<br />

24


II: Mechanistic Approach<br />

Chapter 1: Introduction<br />

Thus far, only a relatively small number <strong>of</strong> studies employing toxicogenomics to study<br />

mechanisms underlying renal carcinogenesis in rats were performed. One study was<br />

designed to identify differential gene expression in kidneys <strong>of</strong> Tsc2 heterozygous mutant<br />

Eker rats compared to wild-type rats, and thereby to elucidate whether those differences<br />

may be involved in tumorigenesis (Sen et al. 2004). The authors found only 3.2% <strong>of</strong> 4395<br />

evaluated genes significantly deregulated, which mainly belonged to the functional<br />

categories <strong>of</strong> cell cycle regulation, cell proliferation, cell adhesion and endocytosis. Notably,<br />

many <strong>of</strong> these genes appear to be directly or indirectly regulated by the PI3K/Akt pathway,<br />

thereby pointing towards mechanistic pathways that may be associated with the enhanced<br />

susceptibility <strong>of</strong> the Tsc2 mutant Eker rats to develop kidney tumors. Prior to this thesis,<br />

gene expression changes in Eker and wild-type rats were not addressed after carcinogen<br />

treatment. It was therefore unknown whether and how genotoxic and non-genotoxic<br />

carcinogens would influence the TSC2 pathway.<br />

Other mechanistic studies used microarrays for the identification <strong>of</strong> deregulated genes and<br />

pathways subsequent to treatment with known genotoxic and non-genotoxic nephrocarcinogens.<br />

For example, Marin-Kuan and colleagues applied microarray technology for<br />

the analysis <strong>of</strong> gene expression changes in kidney samples from F344 rats that were orally<br />

dosed with 300 µg/kg bw OTA during a two year carcinogenicity study. The authors<br />

demonstrated a prominent down regulation <strong>of</strong> genes involved in antioxidant defense, which<br />

was assumed to result in an increase <strong>of</strong> reactive oxygen species, leading to oxidative DNA<br />

damage and tumor initiation (Marin-Kuan et al. 2006). In another study, Chen et al. (Chen<br />

et al. 2006) demonstrated that gene expression pr<strong>of</strong>iling can reveal differential effects <strong>of</strong> AA<br />

exposure on the kidneys and liver <strong>of</strong> rats, i.e. significant alterations <strong>of</strong> genes previously<br />

associated with biotransformation and carcinogenesis, apoptosis and immune response<br />

mainly in kidney, but not in liver, Therefore, in the respective study gene expression<br />

pr<strong>of</strong>iling served to explain underlying mechanisms for the tissue-specific toxicity and<br />

carcinogenicity <strong>of</strong> AA.<br />

These and a number <strong>of</strong> other studies have demonstrated that microarrays facilitate the<br />

interpretation <strong>of</strong> a toxic or carcinogenic mechanism <strong>of</strong> action. The use <strong>of</strong> mechanistic data<br />

in preclinical trials would not only allow to assess differences between acute and chronic<br />

toxicity, but also to extrapolate toxic effects between different species. Moreover,<br />

microarrays were previously used to study gene expression changes in F344 rats<br />

subsequent to treatment with already known non-effective doses and carcinogenic doses <strong>of</strong><br />

potassium bromate. The dramatic increase in global gene expression changes in response<br />

25


Chapter 1: Introduction<br />

to the carcinogenic dose and compared to non-carcinogenic doses suggests that<br />

microarrays are indeed useful tools to determine thresholds and safety levels for test<br />

compounds (Delker et al. 2006).<br />

III: Predictive approach<br />

Histopathology and clinical chemistry subsequent to the two year rodent bioassay are still<br />

benchmarks for evaluating the toxic and carcinogenic potential <strong>of</strong> test compounds.<br />

However, since the kidneys can function relatively normal with low grade damage, altered<br />

blood or urine parameters used for clinical chemistry (e.g. blood urea, nitrogen or serum<br />

creatinine) are not readily observed until a significant portion <strong>of</strong> the kidney is damaged<br />

(Duarte and Preuss 1993). Similarly, histopathologic examinations <strong>of</strong> organ sections require<br />

that toxic lesions (e.g. pre-neoplastic and neoplastic lesions) have become manifested for<br />

microscopic detection. Early or compound-related alterations at a molecular level (e.g. DNA<br />

damage response) thus remain unobserved with these techniques. Microarrays therefore<br />

appear promising for a sensitive prediction <strong>of</strong> toxicity and carcinogenicity.<br />

In general, predictive toxicogenomics require databases with expression pr<strong>of</strong>iles derived<br />

from standardized in vivo or in vitro studies subsequent to treatment with well characterized<br />

pharmaceuticals and chemicals, including time-courses, doses, and clinical pathology<br />

information. These “training pr<strong>of</strong>iles” can then be used to extract sets <strong>of</strong> marker genes to<br />

classify unknown test compounds. Studies employing microarrays for predictions have<br />

been mainly reported for hepatotoxins to extract marker genes representing distinct classes<br />

<strong>of</strong> hepatotoxicity, e.g. liver necrosis or steatosis (Hamadeh et al. 2002; Steiner et al. 2004;<br />

Raghavan et al. 2005). These studies have demonstrated that toxicogenomics can be used<br />

to determine toxicologically relevant changes at very early time points (Steiner et al. 2004)<br />

and at very low doses (Hamadeh et al. 2002). To date, only a few in vivo and in vitro<br />

studies have utilized microarrays to identify early biomarkers that predict chronic endpoints<br />

such as cancer, and that may refine the lifetime rodent bioassay (van Delft et al. 2004;<br />

Tsujimura et al. 2006; Fielden et al. 2007). Most important for this thesis, Ellinger-<br />

Ziegelbauer and colleagues demonstrated that hepatic gene expression pr<strong>of</strong>iles <strong>of</strong> rats<br />

treated for up to 14 days with known genotoxic and non-genotoxic carcinogens allow to<br />

distinguish both groups <strong>of</strong> carcinogens (Ellinger-Ziegelbauer et al. 2005). However, none <strong>of</strong><br />

the available studies dealt with renal cancer.<br />

26


1.5.3 Microarrays- Current Limitations and Future Directions<br />

Chapter 1: Introduction<br />

After several years <strong>of</strong> research, toxicogenomics came into the focus <strong>of</strong> regulatory agencies<br />

and may influence the future decision making on drug safety. Currently, the U.S. Food and<br />

Drug Administration is developing a framework to review gene expression data from<br />

classical toxicological studies (Leighton et al. 2006). However, more research is needed to<br />

adequately assess the potential <strong>of</strong> microarrays for carcinogenicity testing and for the<br />

analysis <strong>of</strong> mechanisms underlying carcinogenesis.<br />

Most <strong>of</strong> the recent studies that analyzed gene expression changes to elucidate mechanism<br />

involved in cancer used whole tumor- or biopsy samples as a source for RNA isolation (Ellis<br />

et al. 2002; Li et al. 2002; Goley et al. 2004; Marin-Kuan et al. 2006; Jones and Pantuck<br />

2008). These samples <strong>of</strong>ten contain different amounts <strong>of</strong> non-cancerous cells, e.g. stromal<br />

or endothelial cells or infiltrating inflammatory cells, thereby complicating an accurate<br />

delineation <strong>of</strong> gene expression changes in non-tumor versus tumor samples. Nevertheless,<br />

the <strong>characteristics</strong> <strong>of</strong> a gross tumor may be influenced by the interaction with these<br />

different, non-cancerous cell types that may contribute to a microenvironment that is<br />

essential for cancer growth, invasion and metastatic progression. Thus it may be relevant<br />

for some, however not for all purposes, to take these cell types under investigation.<br />

In addition, gene expression pr<strong>of</strong>iling seems challenging when analyzing toxic effects in<br />

complex target organs. For instance, the kidney is composed <strong>of</strong> various tubular segments<br />

and cell types, which all have highly specific physiological functions and different<br />

susceptibility towards xenobiotica. Distinct segment-specific nephrotoxic effects may<br />

therefore not be readily detectable by gene expression pr<strong>of</strong>iling when “diluted” by other,<br />

unaffected nephron parts. Separation <strong>of</strong> the kidney into its basal segments (cortex, medulla<br />

and papilla) would serve to highly improve the outcome <strong>of</strong> gene expression experiments.<br />

A major improvement was the development <strong>of</strong> laser capture microdissection (Emmert-Buck<br />

et al. 1996; Schutze et al. 1998), which allows the isolation <strong>of</strong> single cells from a tissue<br />

section, and to collect pure populations <strong>of</strong> morphologically similar cells for subsequent<br />

molecular analysis. Gene expression pr<strong>of</strong>iling from isolated, early preneoplastic lesions<br />

could for instance allow a direct identification <strong>of</strong> gene expression changes underlying<br />

earliest morphologic pathologies. However, previous to this thesis, gene expression<br />

pr<strong>of</strong>iling had not been used for the analysis <strong>of</strong> microdissected pre-neoplastic lesions.<br />

27


Chapter 2: Work Hypotheses & Experimental Design<br />

Lifetime rodent bioassays play a central role in evaluating the carcinogenicity <strong>of</strong> chemicals,<br />

yet are critically viewed especially with regard to their long duration, high dose regimens,<br />

high animal numbers and financial costs. By detecting end-stage adenomas and<br />

carcinomas current protocols enable the identification <strong>of</strong> carcinogens which can act on any<br />

stage <strong>of</strong> tumorigenesis (tumor initiation, promotion and progression). However, they do not<br />

allow drawing detailed conclusions on the underlying molecular mechanisms and their<br />

relevance for human risk assessment. Accordingly, interest in improving carcinogenicity<br />

testing by developing mechanistic and predictive short-term in vivo studies has grown. One<br />

promising approach might be the use <strong>of</strong> microarrays to analyze compound-induced gene<br />

expression changes in different target organs after short-term exposure. However, at the<br />

beginning <strong>of</strong> this thesis:<br />

1. No studies had been performed that elucidated the applicability <strong>of</strong> short-term in vivo<br />

assays in predicting the carcinogenic potential <strong>of</strong> genotoxic and non-genotoxic<br />

renal carcinogens.<br />

2. The identity <strong>of</strong> deregulated genes and pathways involved in the early onset and<br />

progression <strong>of</strong> carcinogen-induced and hereditary renal preneoplastic lesions was<br />

largely unknown.<br />

3. One principal biasing factor for a reliable human risk assessment from rodent<br />

bioassays, i.e. the impact <strong>of</strong> high-fat diet and adiposity on pathways involved in<br />

renal cancer, was never addressed in detail.<br />

2.1 Part I<br />

The first aim <strong>of</strong> this thesis was to investigate the hypothesis that short-term (14 days) in<br />

vivo assays, using microarrays as the detection system, can be used as a tool to evaluate<br />

the renal carcinogenic potential <strong>of</strong> test compounds. Specifically, the most important<br />

prerequisites for short-term in vivo assays to allow for such a sensitive and specific<br />

detection <strong>of</strong> renal carcinogens should be examined, as specified in the below subhypotheses:<br />

28


Chapter 2: Work Hypotheses & Experimental Design<br />

1. Short-term treatment with known renal carcinogens will result in detectable<br />

compound-specific gene expression changes in kidney cortex homogenates <strong>of</strong> rats<br />

(Sensitivity).<br />

2. Gene expression pr<strong>of</strong>iles will serve to distinguish a genotoxic from a non-genotoxic<br />

mode <strong>of</strong> action <strong>of</strong> respective carcinogens (Specificity).<br />

3. Exposure to carcinogens with early genotoxic or non-genotoxic expression changes<br />

in short-tem assays will also result in a carcinogen-specific onset <strong>of</strong> kidney tumors<br />

after subchronic or chronic exposure (Predictivity).<br />

2.1.1 Experimental setup<br />

Short-term exposure: male and female TSC2-mutant Eker rats or wild-type rats were<br />

gavaged daily for 1, 3, 7 and 14 days with the genotoxic carcinogens AA or MAMAc, the<br />

non-genotoxic carcinogen OTA, or vehicle. Subsequent to exposure, kidneys were taken<br />

for the analysis and comparison <strong>of</strong> the following parameters:<br />

• Gene expression pr<strong>of</strong>iles derived from the kidney cortex <strong>of</strong> male Eker and wild-type<br />

rats to detect compound-induced expression changes in both a sensitive and a<br />

conventional rat strain.<br />

• Non-neoplastic and neoplastic renal histopathology in all dose groups to elucidate<br />

specific adverse effects induced by the respective compounds in both rat strains.<br />

• Cell proliferation via immunohistochemical PCNA staining to detect early mitogenic<br />

effects in all dose groups.<br />

• O6-methylguanine DNA adducts levels in 1, 7 and 14 days MAMAc-treated male and<br />

female Eker rats, respectively, indicative for the mutagenic potential <strong>of</strong> MAMAc.<br />

Subchronic and chronic exposure: Male and female Eker rats were gavaged 5 days per<br />

week for 3 or 6 months, respectively, with AA, MAMAc, OTA or vehicle. Subsequently,<br />

kidneys were harvested and analyzed for:<br />

• Sex- and compound-specific differences in non-neoplastic and neoplastic<br />

histopathology (type, incidence and total number <strong>of</strong> lesions).<br />

• Sex- and compound-specific differences in cell proliferation, analyzed by<br />

immunohistochemical BrdU staining.<br />

29


2.2 Part II<br />

Chapter 2: Work Hypotheses & Experimental Design<br />

The second hypothesis <strong>of</strong> this thesis was, that gene expression pr<strong>of</strong>iling from isolated<br />

preneoplastic lesions allows a direct identification <strong>of</strong> gene expression deregulations<br />

underlying earliest morphologic pathologies <strong>of</strong> renal cancer. Specifically the following subhypothesis<br />

should be examined:<br />

1. Different stages <strong>of</strong> preneoplastic lesions can be distinguished by their gene<br />

expression pr<strong>of</strong>iles, therefore allowing the study <strong>of</strong> mechanisms underlying<br />

preneoplastic lesion progression<br />

2. Renal carcinogens differentially affect gene expression preneoplastic lesions,<br />

based on their compound specific mode <strong>of</strong> action (genotoxic vs. non-genotoxic)<br />

3. The comparison <strong>of</strong> gene expression pr<strong>of</strong>iles from short-term treated rats with<br />

microdissected healthy tissue and preneoplastic lesions will serve to identify novel<br />

mechanistic and predictive markers involved in chemical induced renal<br />

carcinogenesis, applicable in short-term assays.<br />

2.2.1 Experimental setup:<br />

Protocol establishment: Before addressing the specific hypotheses, a protocol had to be<br />

established that allows the reliable and reproducible analysis <strong>of</strong> laser microdissected renal<br />

preneoplastic lesions by using Affymetrix Genechips. Specifically, the following conditions<br />

were optimized:<br />

• Effects <strong>of</strong> different histopathological staining techniques in combination with different<br />

sample processing techniques on RNA quantity and quality.<br />

• RNA amplification <strong>of</strong> lowest amounts <strong>of</strong> RNA from dissected lesions to allow reliable<br />

gene expression pr<strong>of</strong>iling.<br />

Microdissection and gene expression analysis: In the next step, the optimized protocol was<br />

adapted to perform large scale gene expression analyses from laser microdissected<br />

lesions:<br />

• Laser microdissection <strong>of</strong> different stages <strong>of</strong> preneoplastic lesions and healthy tubules<br />

from cryosectioned kidneys <strong>of</strong> male Eker rats treated with AA, OTA or vehicle.<br />

30


Chapter 2: Work Hypotheses & Experimental Design<br />

• RNA isolation and microarray analysis <strong>of</strong> the microdissected tissue and comparison<br />

<strong>of</strong> gene expression pr<strong>of</strong>iles between different stages <strong>of</strong> preneoplastic lesions and<br />

healthy tissue.<br />

• Verification <strong>of</strong> deregulated genes, involved in the initiation and progression <strong>of</strong><br />

preneoplastic lesions at the protein level via immunohistochemistry.<br />

2.3 Part III<br />

In the third part <strong>of</strong> this thesis it was hypothesized that chronic high-fat diet causes early<br />

stages <strong>of</strong> renal cancer in rats via mechanisms similar to those observed following exposure<br />

to renal carcinogens. The following sub-hypotheses were addressed:<br />

1. Early stages <strong>of</strong> renal cancer are a direct effect <strong>of</strong> dietary lipids, and largely<br />

independent from the degree <strong>of</strong> body adiposity.<br />

2. Exposure to chronic high fat diet results in the activation the mTOR pathway, a<br />

central component <strong>of</strong> the nutrient-hormonal signaling network and known mediator<br />

<strong>of</strong> renal carcinogenesis.<br />

2.3.1 Experimental setup<br />

From a large pool <strong>of</strong> male Wistar rats fed a customized high-fat diet for 11 months, 2<br />

subgroups <strong>of</strong> diet-induced obesity sensitive and diet-induced obesity resistant rats were<br />

selected, and compared to each other and with low-fat (standard) diet controls for the<br />

following parameters:<br />

• Life history traits (body weight, local fat depots) and plasma parameters (blood<br />

glucose, triglycerides, insulin, leptin, IL-1ß, MCP-1 and PAI-I)<br />

• Possible accumulation <strong>of</strong> renal levels <strong>of</strong> fatty acid binding protein α2u-globulin<br />

• Renal histopathology<br />

• Renal cell proliferation<br />

• Immunodetection <strong>of</strong> hyperphosphorylated S6-ribosomal protein indicative for the<br />

activation <strong>of</strong> the mTOR pathway, a central component <strong>of</strong> the nutrient-hormonal<br />

signaling network and known mediator <strong>of</strong> renal carcinogenesis<br />

31


Chapter 2: Work Hypotheses & Experimental Design<br />

2.4 Schematic Overview on the Experimental Design<br />

Figure 2.1: Overview on the experimental design<br />

32


Chapter 3: Manuscript I<br />

Carcinogen Specific Gene Expression Pr<strong>of</strong>iles in Short-Term<br />

Treated Eker and Wild Type Rats Indicative for Pathways<br />

Involved in Renal Tumorigenesis<br />

Kerstin Stemmer 1 , Heidrun Ellinger-Ziegelbauer 2 , Hans-J. Ahr 2 and Daniel R. Dietrich 1*<br />

1 Human and Environmental Toxicology, University <strong>of</strong> Konstanz, Konstanz, Germany<br />

2 <strong>Molecular</strong> and Special Toxicology, Bayer Healthcare AG, Wuppertal, Germany<br />

* Corresponding author: Daniel R. Dietrich: daniel.dietrich@uni-konstanz.de<br />

Published in <strong>Cancer</strong> Research (2007) May 1;67(9):4052-68<br />

3.1 Abstract<br />

Eker rat heterozygous for a dominant germline mutation in the tuberous sclerosis 2 (Tsc2)<br />

tumor suppressor gene, were used as a model to study renal carcinogenesis. Eker and<br />

corresponding wild type rats were exposed to genotoxic aristolochic acid (AA) or nongenotoxic<br />

ochratoxin A (OTA) in order to elucidate early carcinogen specific gene<br />

expression changes and to test whether Eker rats are more sensitive for carcinogen<br />

induced changes in gene expression. Male Eker and wild type rats were gavaged daily with<br />

AA (10mg/kg BW) or OTA (210µg/kg BW). After 1, 3, 7 and 14 days <strong>of</strong> exposure, renal<br />

histopathology, tubular cell proliferation and Affymetrix gene expression pr<strong>of</strong>iles from renal<br />

cortex/outer medulla were analyzed. AA-treated Eker and wild type rats were qualitatively<br />

comparable in all parameters assessed, suggesting a Tsc2-independent mechanism <strong>of</strong><br />

action. OTA treatment resulted in slightly increased cortical pathology and significantly<br />

elevated cell proliferation in both strains, although Eker rats were more sensitive.<br />

Deregulated genes involved in PI3K-AKT-Tsc2-mTor signalling, amongst other important<br />

genes prominent in tumorigenesis, in conjunction with the enhanced cell proliferation and<br />

presence <strong>of</strong> preneoplastic lesions suggested involvement <strong>of</strong> Tsc2 in OTA- mediated toxicity<br />

33


Chapter 3: Manuscript I<br />

and carcinogenicity, especially as deregulation <strong>of</strong> genes involved in this pathway was more<br />

prominent in the Tsc2 mutant Eker rat.<br />

3.2 Introduction<br />

Eker rats, heterozygous for a loss-<strong>of</strong>-function mutation in the tuberous sclerosis 2 (Tsc2)<br />

tumor suppressor gene, appear ideal models to study the aetiology <strong>of</strong> renal carcinogenesis<br />

(Yeung et al. 1994; Hino et al. 1995). Heredity <strong>of</strong> the Tsc2 mutation follows Mendelian<br />

genetics (Yeung et al. 1994; Kobayashi et al. 1995) and heterozygous progeny are<br />

predisposed to spontaneous development <strong>of</strong> multiple bilateral renal neoplasms originating<br />

from the proximal tubular epithelium with complete penetrance by one year <strong>of</strong> age (Everitt<br />

et al. 1995). Approximately 60% <strong>of</strong> the spontaneous renal tumors in Eker rats also exhibit a<br />

functional inactivation <strong>of</strong> the second Tsc2 allele, suggesting, in accordance with Knudson’s<br />

two-hit hypothesis that a second somatic mutation might be the rate-limiting step for the<br />

development <strong>of</strong> renal cell carcinomas (RCC) in Eker rats (Yeung et al. 1995). Eker rats<br />

have been employed to elucidate the mechanism <strong>of</strong> renal carcinogens, primarily using<br />

histopathological and statistical analyses <strong>of</strong> the number, multiplicity and progression <strong>of</strong><br />

renal lesions (Walker et al. 1992; Wolf et al. 2000). Accordingly, treatment <strong>of</strong> Eker rats with<br />

dimethylnitrosamine resulted in a 70-fold increase in the induction <strong>of</strong> renal adenomas and<br />

carcinomas, when compared to wild type rats (Walker et al. 1992). No increased lesion<br />

incidence, albeit an advanced lesion progression was observed in Eker rats, subchronically<br />

treated with the tumor promoter sodium barbital (Wolf et al. 2000). Although the latter data<br />

highlight that Eker rats are sensitive to genotoxic and non-genotoxic compounds, the<br />

involvement <strong>of</strong> Tsc2 protein (tuberin) in renal carcinogenesis remains to be established.<br />

Several studies suggest that functional Tsc2 promotes the GTP-hydrolysis <strong>of</strong> the Ras<br />

homologe Rheb, thereby acting as a negative regulator <strong>of</strong> the PI3K-Akt-Tsc1/2-Rheb-mTor<br />

pathway. Consequently, Tsc2 is suspected to play a central role in mediating growth factor,<br />

nutrient and energy sensing to regulate cell growth, proliferation, migration and<br />

differentiation (Mak and Yeung 2004).<br />

The objective <strong>of</strong> this study was to elucidate whether short-term exposure <strong>of</strong> Eker and wild<br />

type rats to a non-genotoxic and a genotoxic renal carcinogen would result in compound<br />

specific changes in renal non-neoplastic and preneoplastic pathology and cell proliferation<br />

rates. Subsequently the hypothesis was investigated, whether compound specific changes<br />

34


Chapter 3: Manuscript I<br />

in histopathology and cell proliferation can be associated with respective changes in gene<br />

expression and whether Eker and wild type rats respond differently. This should allow<br />

identification <strong>of</strong> deregulated genes involved in known and novel pathways possibly<br />

mediating carcinogen-induced renal tumorigenesis. Accordingly, Eker and wild type rats<br />

were treated with daily doses <strong>of</strong> the genotoxic and the non-genotoxic renal carcinogen<br />

aristolochic acid (AA) and ochratoxin A (OTA), respectively, for which renal tumor induction<br />

in long-term in vivo studies was previously demonstrated (Mengs et al. 1982; Boorman et<br />

al. 1992).<br />

Indeed, intragastric administration <strong>of</strong> 10mg AA /kg BW/day (representing a mixture <strong>of</strong><br />

structurally related nitrophenanthrene carboxylic acids (mostly AAI and AAII)) to rats over<br />

three months was demonstrated to induce tumors in the forestomach, kidney and the<br />

urinary bladder (Mengs et al. 1982). DNA reactivity <strong>of</strong> AA was confirmed in that the most<br />

frequent and persistent dAdenin- AAI adduct could lead to mutation and activation <strong>of</strong> the Hras<br />

oncogene in the forestomach but not in kidneys <strong>of</strong> rats (Schmeiser et al. 1991; Cheng<br />

et al. 2006) or to p53-mutations in urothelial tumors <strong>of</strong> humans (Lord et al. 2004). Despite<br />

the lack <strong>of</strong> H-ras mutations, higher levels <strong>of</strong> AA adducts were found in renal tissues than in<br />

the forestomach <strong>of</strong> orally treated Wistar rats (5mg AA/kg BW/day) after only one week <strong>of</strong><br />

exposure (Dong et al. 2006), suggesting an H-ras independent pathway <strong>of</strong> renal tumor<br />

induction. The genotoxic properties <strong>of</strong> AA are explained by the metabolic activation <strong>of</strong> AA<br />

by several phase I enzymes to a DNA-reactive aristolactam-nitriumion (Pfau et al. 1990;<br />

Arlt et al. 2002; Stiborova et al. 2003). Similarly, the mycotoxin OTA increased the<br />

incidence <strong>of</strong> renal adenoma and carcinoma in rats, when exposed for up to two years to<br />

dietary OTA (Mantle et al. 2005) or 210µg OTA /kg BW/day via gavage (Boorman et al.<br />

1992). However, as OTA has not been convincingly demonstrated to covalently interact<br />

with DNA, a non-genotoxic mechanism <strong>of</strong> action is assumed (Kamp et al. 2005; Mally et al.<br />

2005).<br />

The comparison <strong>of</strong> cell proliferation, pathology and expression pr<strong>of</strong>iles <strong>of</strong> AA- and OTAtreated<br />

Eker and wild type rats should allow for a more in-depth understanding <strong>of</strong> the<br />

involvement <strong>of</strong> the Tsc2-mTor pathway as well as <strong>of</strong> other early gene expression changes<br />

in the etiology <strong>of</strong> carcinogen induced renal tumors.<br />

35


3.3 Material and Methods<br />

3.3.1 Compounds<br />

Chapter 3: Manuscript I<br />

Ochratoxin A (>98% purity, benzene free) was kindly provided by Dr. M.E. Stack, US FDA,<br />

Washington DC. Aristolochic acid sodium salt mixture (AA I: 41% and AA II: 56%) was<br />

purchased from Sigma Aldrich, Germany.<br />

3.3.2 Animals<br />

Six to ten week old, genotyped heterozygous Tsc2 mutant Eker rats (Tsc2+/-, Long Evans)<br />

were purchased from the MD Anderson <strong>Cancer</strong> Center, Smithville, Texas, USA and<br />

maintained at the University <strong>of</strong> Konstanz animal research facility under standard conditions<br />

with food and water ad libitum. Male rats were randomly assigned to dose groups (three<br />

animals per compound (or vehicle) and time-point) and allowed to acclimatize to laboratory<br />

conditions for 4 weeks. Two weeks prior to exposure, rats were handled daily to reduce<br />

non-compound related stress during exposure.<br />

Heterozygous Eker rats were bred and wild type (Tsc2+/+) genotypes <strong>of</strong> the progeny were<br />

determined via PCR (Rennebeck et al. 1998). Two weeks prior to exposure, 8-9 week old<br />

genotyped male wild type rats, were randomly allocated to dose groups and accustomed to<br />

daily handling (see above).<br />

3.3.3 Animal treatment and sample collection<br />

Eker and wild type rats were gavaged daily with OTA (210µg/ kg BW) or AA (10mg/ kg<br />

BW), dissolved in 0,1M sodium bicarbonate. Time-matched vehicle controls were gavaged<br />

with 0,1M sodium bicarbonate. Following 1, 3, 7, and 14 days <strong>of</strong> treatment, Narcoren®<br />

(pentobarbital) anesthetized rats were sacrificed by exsanguination subsequent to<br />

retrograde perfusion with PBS. Left kidneys were collected, cross-sectioned into 5mm<br />

slices and stored in RNAlater (Qiagen, Germany) or in PBS buffered histology fixative<br />

buffer, containing 2% paraformaldehyde and 1% glutaraldehyde for subsequent paraffin<br />

embedding and sectioning.<br />

36


3.3.4 Histopathology<br />

Chapter 3: Manuscript I<br />

For histopathological examinations, H&E stained sections were randomized and<br />

pathological analysis was carried out by light microscopy at 40-400-fold magnification. Nonneoplastic<br />

changes were classified as none (0), mild (1), moderate (2), strong (3), and<br />

severe (4), including intermediate classes (e.g. 0.5, 1.5 etc.), while total numbers <strong>of</strong><br />

preneoplastic and neoplastic lesions were counted.<br />

3.3.5 Immunohistochemistry<br />

Cell proliferation was evaluated by immunohistochemical staining for proliferating cell<br />

nuclear antigen (PCNA) using monoclonal primary anti-PCNA antibody (PC-10; DAKO,<br />

Germany) in paraffin-embedded kidney sections.<br />

Sections were deparaffinized, rehydrated in a decreasing alcohol series and washed with<br />

PBS. For antigen retrieval, slides were placed in 0,1M sodium citrate buffer (pH 6.0),<br />

microwaved to boiling point 3-times and cooled to room temperature for 20min. Sections<br />

were denatured with 4 N HCl (20min at 37°C), washed with PBS (2x 5min) and non-specific<br />

protein binding blocked by preincubation with casein solution (Power BlockTM, BioGenex,<br />

USA) for 20 minutes. Sections were incubated with PC-10 primary antibody (diluted 1:50 in<br />

Power BlockTM) at 4°C for 16 hours. Antigen- antiserum complexes were visualised using<br />

the Super SensitiveTM alkaline phosphatase labelled, biotin streptavidine amplified<br />

detection system and Fast Red as chromogen according to the manufacturer’s instructions<br />

(BioGenex, USA).<br />

Cell proliferation was quantified on PCNA stained sections, randomized across all<br />

treatment and control groups. Twenty microscopic fields (10x ocular, 40x objective) were<br />

randomly chosen in the outer cortex and inner cortex/outer medulla. All tubule cell nuclei<br />

were counted, concurrently differentiating between negative and positive PCNA staining.<br />

Nuclear labeling indices for PCNA (LI%) (PCNA positive nuclei/total number <strong>of</strong> nuclei<br />

counted) were determined based on a minimum <strong>of</strong> at least 2.000 nuclei evaluated.<br />

3.3.6 RNA isolation and expression pr<strong>of</strong>iling<br />

RNA isolation from RNAlater fixed kidneys was performed as described previously<br />

(Ellinger-Ziegelbauer et al. 2005). Starting with 5µg <strong>of</strong> total RNA with a 28S/18S rRNA peak<br />

37


Chapter 3: Manuscript I<br />

ratio >1.7, biotin-labeled cRNA was prepared and subsequently hybridized on Affymetrix<br />

Rat Genome RAE230A arrays according to the manufacturer’s instructions (Affymetrix,<br />

USA; GeneChip® Expression Analysis 701194 Rev.1). This specific array contains 15.866<br />

probe sets, corresponding to approx. 5399 annotated rat genes and 10467 expressedsequence<br />

tags (ESTs).<br />

3.3.7 Microarray data processing and statistical analysis<br />

Microarray quality control was performed as described previously (Ellinger-Ziegelbauer et<br />

al. 2005) and gene expression data were submitted to the GEO repository<br />

(http://www.ncbi.nlm.nih.gov/projects/geo; accession number: GSE5923). Expressionist<br />

Analyst s<strong>of</strong>tware (Genedata AG, Switzerland) was used for statistical analysis. Significantly<br />

deregulated genes per compound were selected based on the factors treatment and time<br />

as both, single and interaction effects, in a 2-way ANOVA with a p-value cut-<strong>of</strong>f <strong>of</strong> 0.005,<br />

combined with a 1.7-fold deregulation threshold for at least one time point. Significantly<br />

deregulated genes were divided into gene groups with distinct expression pr<strong>of</strong>iles over the<br />

time-course using self-organizing map (SOM) analysis. SOM analysis also allowed<br />

deselection <strong>of</strong> genes showing inconsistent expression between the controls at different time<br />

points. Using the adjusted datasets, gene expression ratios <strong>of</strong> individual genes were<br />

calculated by dividing the respective expression values <strong>of</strong> single treated replicate samples<br />

by the mean expression value <strong>of</strong> all corresponding time-matched control samples.<br />

Heatmaps were used to graphically display the relative expression data, after onedimensional<br />

clustering <strong>of</strong> the genes (for the validation <strong>of</strong> microarray data, see<br />

supplementary information).<br />

3.3.8 Functional analysis <strong>of</strong> microarray data<br />

For functional analysis each significantly deregulated gene was characterized according to<br />

the biochemical role <strong>of</strong> its encoded protein, whenever sufficient information from<br />

databases, e.g. NetAffx from Affymetrix (update from August 2006), Swissprot, Proteome<br />

and Pubmed was available. The consequence <strong>of</strong> the direction <strong>of</strong> deregulation was<br />

interpreted specifically with regard to possible downstream pathophysiological effects. This<br />

allowed distribution <strong>of</strong> the deregulated genes into toxicological categories (Supplementary<br />

table 3.1) and facilitated the comparison <strong>of</strong> specific pathophysiological pathways, involved<br />

38


Chapter 3: Manuscript I<br />

in the response <strong>of</strong> Eker and wildy type rats to AA and OTA treatment. In addition, the<br />

pathophysiological pathways were compared with major pathways suspected to be<br />

involved in AA- and OTA- induced carcinogenesis (Tables 3.1 and 3.2) and<br />

histopathological changes observed.<br />

3.3.9 Statistical analysis<br />

Statistical analysis <strong>of</strong> histopathological and cell proliferation changes were carried out using<br />

GraphPad Prism 4® (USA) S<strong>of</strong>tware. Statistical significant differences in nuclear labeling<br />

indices (LI %) or total number <strong>of</strong> lesions in treated and control animals were analyzed by an<br />

unpaired (two tailed) t-test. A statistical significant effect <strong>of</strong> the treatment- time response<br />

was tested with a 2-Way ANOVA.<br />

3.4 Results<br />

3.4.1 Cell proliferation data<br />

AA treatment resulted in no overt change in cell proliferation rate (Figure 3.1, A and B) in<br />

either strain, despite that occasional AA groups appeared to have a significantly lower<br />

proliferation then the corresponding controls. Conversely, OTA treatment increased the<br />

proliferation rate on day 14 (Figure 3.1, C and D, Figure 3.2, A and B) 3.8- and 3.4-fold<br />

above control in Eker and wild type rats, respectively. Moreover, Eker rats appeared to be<br />

more sensitive to OTA as an increased cell proliferation rate was already observed at day 7<br />

<strong>of</strong> treatment.<br />

3.4.2 Preneoplastic and neoplastic pathology<br />

While no preneoplastic or neoplastic lesions (Dietrich and Swenberg 1991) were observed<br />

in any <strong>of</strong> the wild type rat controls, the Eker rat control groups presented with atypical<br />

tubules (AT, Figure 3.2, C), atypical hyperplasia (AH, Figure 3.2, D) and an occasional<br />

adenoma (Supplementary table 3.2). Neither AA nor OTA treatment induced an increase in<br />

preneoplastic or neoplastic lesions in wild type rats. Similarly, AA treatment <strong>of</strong> Eker rats<br />

39


Chapter 3: Manuscript I<br />

resulted in no significant increase in AT or AH. However, two carcinomas were observed in<br />

the day 14 AA-treated Eker group, and an adenoma in the day one group. In contrast, OTA<br />

treatment <strong>of</strong> Eker rats resulted in a significant increase <strong>of</strong> AT on day 14 (Figure 3.1, E).<br />

However, no significant increase in AH or neoplastic lesions were observed.<br />

Figure 3.1: Comparison <strong>of</strong> PCNA S-Phase labelling indices (LI %) in the renal cortex <strong>of</strong> AA or<br />

OTA treated Eker and wild type rats (A-D), respectively, at various treatment timepoints.<br />

E: Mean number <strong>of</strong> atypical tubules (preneoplastic lesion) per animal in<br />

control and OTA treated Eker rats. Data represent the mean + SEM <strong>of</strong> n=3 animals per<br />

group. Significant differences between treated and time-matched control groups: *p <<br />

0.05; **p < 0.01 and ***p


Chapter 3: Manuscript I<br />

Figure 3.2: Representative PCNA staining patterns <strong>of</strong> control- (A) and OTA-treated (B) Eker rats.<br />

C: Atypical tubule; D: Atypical hyperplasia; E: karyomegally (arrows) and F: apoptotic<br />

nuclei (arrow) observed in OTA treated rats.<br />

41


3.4.4 Gene expression pr<strong>of</strong>iles<br />

Chapter 3: Manuscript I<br />

Oral treatment <strong>of</strong> Eker and wild type rats with AA and OTA, respectively, led to a significant<br />

deregulation <strong>of</strong> gene expression already after one day <strong>of</strong> exposure. Compared to the<br />

respective time-matched controls, the number <strong>of</strong> significantly deregulated genes increased<br />

with the duration <strong>of</strong> exposure in both strains. At all time-points compound-treated Eker rats<br />

consistently showed a higher number <strong>of</strong> significantly deregulated genes compared to their<br />

wild type counterparts (Figure 3.3). Overall, AA treatment led to the significant deregulation<br />

<strong>of</strong> 111 non-redundant genes in Eker rats compared to 81 genes in wild type rats. However,<br />

the visualization <strong>of</strong> the expression pr<strong>of</strong>iles <strong>of</strong> the union <strong>of</strong> the Eker- and wild type- selected<br />

genes revealed a qualitatively comparable pr<strong>of</strong>ile in AA-treated Eker and wild type rats. In<br />

contrast, treatment with OTA resulted in 375 significantly deregulated, non-redundant<br />

genes in Eker rats compared to 141 genes in wild type rats, with a strikingly different<br />

expression pr<strong>of</strong>ile <strong>of</strong> at least half <strong>of</strong> the genes (Figure 3.3).<br />

Figure 3.3: Heat map <strong>of</strong> compound-specific unions <strong>of</strong> genes found significantly deregulated (red:<br />

upregulated; green: downregulated) from the corresponding control in AA or OTA<br />

induced renal gene expression pr<strong>of</strong>iles <strong>of</strong> Eker or wild type rats after 1, 3, 7 or 14 days<br />

<strong>of</strong> treatment (n=3 per time-point). Color scale (left side): gene expression ratio.<br />

42


3.4.5 Functional analysis <strong>of</strong> significantly deregulated genes<br />

Chapter 3: Manuscript I<br />

Many <strong>of</strong> the non-redundant genes, deregulated by AA (Table 3.1) or OTA (Table 3.2)<br />

treatment in Eker and wild type rats, respectively, could be associated with major<br />

pathophysiological processes involved in renal toxicity and regeneration (Supplementary<br />

table 3.1).<br />

3.4.6 Genes deregulated by aristolochic acid<br />

Metabolism and bioactivation: Treatment <strong>of</strong> Eker and wild type rats with AA led to a<br />

prominent up-regulation <strong>of</strong> genes encoding phase I or phase II biotransformation enzymes<br />

or drug transporters. Most <strong>of</strong> the genes were either constantly upregulated over the whole<br />

exposure time-frame or increasingly upregulated with prolonged duration <strong>of</strong> exposure in<br />

both Eker and wild type rats. In addition to the genes significantly deregulated in both<br />

strains, some genes met the significance criteria only in one or the other strain. Yet the<br />

direction <strong>of</strong> deregulation was mostly comparable (e.g. GSTM2 or CES3, apparently specific<br />

for Eker or wild type rats, respectively).<br />

DNA damage response (incl. oxidative stress): P53 is inducible by DNA damage and<br />

oxidative stress. The upregulation <strong>of</strong> several p53 pathway genes, as observed in Eker and<br />

wild type rats, was therefore summarized as DNA damage response (incl. oxidative stress).<br />

Most genes involved in this category showed a time-dependent increase with highest<br />

deregulation values after 14 days <strong>of</strong> treatment and met the significance criteria in both<br />

strains.<br />

Inhibited cell survival and proliferation: In conjunction with the DNA damage response<br />

described above, downregulation <strong>of</strong> anti-apoptotic genes and genes involved in DNA<br />

replication, cell cycle progression, as well as the upregulation <strong>of</strong> pro-apoptotic genes was<br />

observed. These genes demonstrated a comparable time-dependent increase in<br />

expression as the genes representing the DNA damage response (see above), with highest<br />

deregulation values after 14 days <strong>of</strong> treatment. In contrast to the DNA damage response<br />

genes, many <strong>of</strong> these genes showed an apparently strain-specific deregulation, yet again<br />

with a qualitatively similar expression pattern for most <strong>of</strong> them in both strains. An exception<br />

to the latter observation, were genes directly involved in the G2/M transition <strong>of</strong> the cell<br />

cycle, which were specifically and consistently downregulated in wild type rats.<br />

43


Chapter 3: Manuscript I<br />

Enhanced cell survivial/cell proliferation: Only three genes were assigned to this<br />

category. The Tsc1 (tuberous sclerosis 1) tumor suppressor gene, which is known to be<br />

associated with Tsc2 (Mak and Yeung 2004), was significantly downregulated in Eker but<br />

not wild type rat. Enhanced cell survival and proliferation induced by AA was suggested by<br />

the observed upregulation <strong>of</strong> a positive regulator <strong>of</strong> cell proliferation (KEG1) and the<br />

downregulation <strong>of</strong> a proapoptotic gene (WWOX).<br />

44


Chapter 3: Manuscript I<br />

Table 3.1: Categories <strong>of</strong> genes differentially deregulated by AA in Eker- and wild type rats. For the major toxicological categories the associated genes<br />

are listed together with their Genebank accession number. The main biochemical functions or pathways in which these genes are involved are<br />

indicated in column four. For Eker- and wild type rats, the fold deregulation ratios <strong>of</strong> genes that were significantly deregulated over all timepoints<br />

according to N-way ANOVA are shown in the last eight columns. Genes meeting the significance criteria are indicated in bold.<br />

Toxicological<br />

category<br />

Metabolism and<br />

Biotransformation<br />

Accession<br />

number<br />

Gene title Biochemical category Fold deregulation (mean <strong>of</strong> three replicate animals<br />

per time-point)<br />

AI233740 AKR1B8 Aldo-keto reductase 1B8 Biotransformation, Phase I 2.2 3.4 2.5 23.3 1.8 2.0 2.7 2.7<br />

NM_012844 EPHX1 Epoxide hydrolase 1 (microsomal) Biotransformation, Phase I 1.7 1.7 1.5 1.9 1.2 1.3 1.4 1.8<br />

AW142784 CYP4A10 Cytochrome P450 4a10 Biotransformation, Phase I 2.0 1.6 1.6 2.1 1.6 1.8 1.9 1.8<br />

J02679 NQO1 NADPH-Quinone Oxidoreductase Biotransformation, Phase I 2.4 1.9 1.6 3.1 2.0 2.1 1.5 2.0<br />

U27518 UGT2B8 UDP-glucuronosyltransferase 2B8, micros. Biotransformation, Phase II 6.1 4.5 3.3 4.7 3.1 2.5 1.7 1.7<br />

NM_031980 UGT2B12 UDP-glucuronosyltransferase 2B12, micros. Biotransformation, Phase II 2.0 2.1 1.9 2.2 2.2 2.0 2.0 1.9<br />

M31109 UGT2B3 UDP-glucuronosyltransferase 2B3, micros. Biotransformation, Phase II 4.1 4.6 4.5 6.7 2.7 2.0 4.7 3.6<br />

M28241 GSTM1 Glutathione-S-transferase M1 Biotransformation, Phase II 4.2 5.5 5.4 15.8 3.9 3.1 3.6 7.1<br />

NM_031154 GSTM3 Glutathione-S-transferase M3 Biotransformation, Phase II 1.1 2.0 1.6 2.0 1.3 1.3 1.4 2.1<br />

NM_053293 GSTT1 Glutathione-S-transferase T1 Biotransformation, Phase II 1.3 1.6 2.0 2.0 1.3 1.3 1.6 1.8<br />

AA945082 GSTA2 Glutathione-S-transferase A2 Biotransformation, Phase II 4.1 4.5 2.9 3.8 3.9 4.3 4.2 5.0<br />

X02904 GSTP1 Glutathione-S-transferase P1 Biotransformation, Phase II 4.8 5.7 3.9 4.8 4.9 4.9 5.3 6.0<br />

NM_013215 AKR7A3 Aldo-keto reductase family 7, member A3 Biotransformation, Phase I 1.8 1.7 1.5 2.0 1.5 1.4 1.5 1.7<br />

NM_017084 GNMT Glycine-N-methyltransferase Biotransformation, Phase I 1.4 1.6 1.8 1.6 1.2 1.2 1.0 1.9<br />

NM_133586 CES2 Carboxylesterase 2 (intestine, liver) Biotransformation, Phase I -1.1 1.1 1.1 2.1 -1.4 1.1 1.5 2.9<br />

AW142784 CYP51 Cytochrome P450 51 Biotransformation, Phase I 1.1 -1.4 -1.7 -1.1 1.0 1.0 -1.6 -1.1<br />

BI285792 GSTM7-7 Similar to glutathione transferase GSTM7-7 Biotransformation, Phase II 1.5 1.7 1.5 2.1 1.3 1.5 1.3 1.5<br />

AI169331 GSTM2 Glutathione-S-transferase M2 Biotransformation, Phase II 1.1 1.2 1.7 2.1 1.0 1.2 1.3 1.9<br />

AF461738 UGT1A UDP-glucuronosyltransferase 1A, micros. Biotransformation, Phase II 1.9 1.7 1.5 1.8 1.5 1.6 1.6 1.5<br />

Ek<br />

d1<br />

Ek<br />

d3<br />

Ek<br />

d7<br />

Ek<br />

d14<br />

wt<br />

d1<br />

wt<br />

d3<br />

wt<br />

d7<br />

wt<br />

d14<br />

45


DNA damage<br />

response (incl.<br />

oxidative stress<br />

response)<br />

Inhibited cell<br />

survival and<br />

proliferation<br />

Accession<br />

number<br />

Chapter 3: Manuscript I<br />

Gene title Biochemical category Fold deregulation (mean <strong>of</strong> three replicate animals<br />

per time-point)<br />

NM_133547 SULT1C2 Sulfotransferase K1 (SULTK1) Biotransformation, Phase II -1.3 -1.2 -1.3 -2.0 1.0 -1.3 -1.1 -1.3<br />

AY082609 MDR1B Mutlidrug resistance-1B (ABCB1b) Drug transport 1.1 1.2 1.2 5.5 -1,1 1.1 1.6 2.5<br />

NM_012833 MRP2 Multidrug resistance-associated protein 2 Drug transport 1.5 1.5 1.2 4.6 1.1 1.3 1.1 1.6<br />

L46791 CES3 Carboxylesterase 3 Biotransformation, Phase I 1.1 1.5 1.5 1.7 1.4 1.6 1.6 2.3<br />

NM_134349 MGST1 Microsomal glutathione S-transferase 1 Biotransformation, Phase II 1.5 1.4 1.3 1.5 1.6 1.4 1.5 1.8<br />

AF072816 MRP3 Multidrug resistance-associated protein 3 Drug transport 4.1 -1.7 1.3 2.2 2.3 1.3 1.9 7.8<br />

NM_012861 MGMT O6-methylguanine-DNA methyltranferase DNA repair -1.1 1.2 1.7 3.1 1.1 1.2 1.9 2.7<br />

AW520812 PHLDA3 Pleckstrin homology-like domain family A3 DNA damage response 1.1 1.1 1.3 3.2 1.4 1.4 2.4 3.2<br />

Q64315 CDKN1A Cyclin-dependent kinase inhibitor 1A (p21) Cell cycle checkpoint 1.5 1.1 6.9 14.2 1.1 2.7 2.8 9.8<br />

NM_031821 SNK Serum-inducible kinase (PLK2) Cell cycle checkpoint 1.2 1.3 2.2 6.6 1.4 1.2 2.0 4.3<br />

NM_012923 CCNG1 Cyclin G1 Cell cycle checkpoint 1.1 1.5 2.3 3.8 1.1 1.6 2.1 4.1<br />

BI296301 MDM2 Ubiquitin E3 ligase Mdm2 (predicted) Cell cycle checkpoint 1.0 1.7 1.4 4.9 -1.2 1.0 1.9 2.4<br />

NM_022547 FTHFD 10-formyltetrahydr<strong>of</strong>olate dehydrogenase Cell cycle 2.0 1.9 1.5 1.8 1.7 1.7 1.6 1.8<br />

AI411345 PRODH Similar to Proline dehydrogenase (oxidase) 1 Proapoptotic 1.4 2.5 1.7 2.0 1.2 1.7 1.8 1.9<br />

NM_057153 OXR1 Oxidation resistance 1 DNA damage response -1.1 1.2 1.6 1.5 1.2 -1.1 1.2 1.7<br />

AA801395 TNFAIP8 Tumor necrosis factor α-induced protein 8 Antiapoptotic 1.1 -1.3 -1.4 -2.5 1.0 1.1 1.2 -1.4<br />

M14050 HSPA5 Heat shock 70kD protein 5 (GRP78) Antiapoptotic 1.5 -1.7 -1.6 -1.4 1.0 1.2 1.1 -1.3<br />

AF106659 USP2 Ubiquitin-specific protease 2 Proapoptotic -1.1 1.9 1.6 3.5 1.0 2.0 1.5 1.4<br />

BF288101 SNN Stannin Proapoptotic -1.2 1.1 1.1 1.9 -1.1 1.1 1.0 1.8<br />

AI714002 Ki-67 (predicted) DNA replication -2.4 -2.2 -1.9 -1.2 1.5 -1.4 1.0 -1.4<br />

AI072892 FRZB Frizzled-related protein Signalling cascades -1.1 1.2 1.8 1.3 1.6 1.2 1.0 1.3<br />

M15481 IGF1 Insulin-like growth factor 1 Regulation <strong>of</strong> proliferation -1.1 -1.2 -1.7 -2.4 1.0 1.2 -1.1 -2.0<br />

NM_022266 CTGF Connective tissue growth factor Cell adhesion / migration 1.2 -1.1 -1.1 -1.9 1.4 1.1 1.2 -1.9<br />

Ek<br />

d1<br />

Ek<br />

d3<br />

Ek<br />

d7<br />

Ek<br />

d14<br />

wt<br />

d1<br />

wt<br />

d3<br />

wt<br />

d7<br />

wt<br />

d14<br />

46


Enhanced cell<br />

survival/<br />

proliferation<br />

Accession<br />

number<br />

Chapter 3: Manuscript I<br />

Gene title Biochemical category Fold deregulation (mean <strong>of</strong> three replicate animals<br />

per time-point)<br />

BI275994 TGM2 Tissue-type transglutaminase Cell adhesion / migration -1.7 -1.5 -1.5 -2.9 -1.9 -2.0 -1.9 -1.9<br />

AI102530 NAB2 Ngfi-A binding protein 2 (predicted) Transcriptional corepressor -1.1 1.2 1.8 2.0 1.1 1.0 1.0 1.7<br />

BF417638 CDCA3 Cell division cycle associated 3 (TOME1) Cell cycle (G2/M) 1.1 -1.1 -1.3 1.3 -1.2 -1.7 -1.6 -1.5<br />

X64589 CCNB1 Cyclin B1, G2/M-specific Cell cycle (G2/M) 1.0 -1.3 -1.3 1.0 -1.4 -2.2 -1.3 -1.6<br />

AI171185 HMMR Hyaluronan mediated motility receptor Cell cycle (G2/M) 1.2 -1.4 -1.4 1.2 -1.6 -2.0 -1.4 -1.7<br />

NM_019296 CDC2 Cell division cycle 2 protein kinase (CDK1 Cyclin<br />

dependent kinase 1)<br />

Ek<br />

d1<br />

Ek<br />

d3<br />

Ek<br />

d7<br />

Ek<br />

d14<br />

Cell cycle (G2/M) -1.2 -1.4 -2.5 1.2 -1.5 -2.4 -2.0 -1.6<br />

AA944180 CKS2 Cyclin-dependent kinases regulatory subunit 2 Cell cycle (G2/M) 1.0 1.1 -1.7 2.5 -1.6 -2.3 -2.6 -1.3<br />

BF396602 SFRP2 secreted frizzled-related protein 2 Antiapoptotic -1.1 -1.2 -1.6 -1.4 -1.1 -1.1 -1.4 -1.7<br />

NM_012593 KLK7 Kallikrein 7 Antiapoptotic 1,2 -1.3 -1.8 -2.2 -1.4 -1.1 -1.2 -1.8<br />

NM_021854 TSC1 Tuberous sclerosis 1 Tumor suppressor gene -1.9 -2.1 -1.3 -2.3 -1.1 -1.8 1.0 -1.5<br />

BE098555 WWOX WW-domain oxidoreductase (predicted) Proapoptotic -1.4 -1.5 -1.5 -1.7 -1.5 -1.5 -1.9 -1.9<br />

NM_134330 KEG1 kidney expressed gene 1 Regulation <strong>of</strong> proliferation 1.2 1.0 1.8 1.3 1.7 1.2 1.8 2.2<br />

wt<br />

d1<br />

wt<br />

d3<br />

wt<br />

d7<br />

wt<br />

d14<br />

47


3.4.7 Genes deregulated by ochratoxin A<br />

Chapter 3: Manuscript I<br />

Biotransformation: OTA treatment downregulated the expression <strong>of</strong> several phase I and<br />

phase II enzymes and drug transporter genes in both strains. While this effect appeared<br />

constant over time for some genes, most deregulated genes demonstrated an enhanced<br />

downregulation with increasing exposure time. Besides the downregulation <strong>of</strong> numerous<br />

genes coding for components <strong>of</strong> the biotransformation machinery, OTA treatment resulted<br />

in an increased upregulation <strong>of</strong> the phase I gene CYP4A12 in both strains and a consistent<br />

upregulation <strong>of</strong> phase I ALDH6A1 in Eker rats.<br />

DNA damage response (incl. oxidative stress): OTA treatment led to significant<br />

upregulation <strong>of</strong> p53 pathway genes in both strains. However, the upregulation <strong>of</strong> these<br />

genes differed between the two strains. Eker rather than wild type rats demonstrated an<br />

upregulation <strong>of</strong> genes known to be involved in oxidative stress responses as well as<br />

downregulation <strong>of</strong> genes that code for products with extra-cellular antioxidant activities. The<br />

latter genes and the upregulated KEAP1, which suppresses the transactivation <strong>of</strong><br />

antioxidant responsive elements, were categorized as “enhanced oxidative stress”.<br />

Cellular stress: A general stress response, as indicated primarily by the upregulation <strong>of</strong><br />

several components <strong>of</strong> the stress-inducible MAP-kinase pathway, was predominantly<br />

detectable in Eker rats, since the latter genes were not significantly deregulated in wild type<br />

rats.<br />

Inhibited cell survival and proliferation: Predominantly OTA-treated Eker rats presented<br />

with an upregulated expression <strong>of</strong> tumor suppressor-, negative cell proliferation controland<br />

pro-apoptotic response genes. However, in both strains an inhibited cell survival<br />

response could be inferred from the upregulation <strong>of</strong> a pro-apoptotic gene and the<br />

downregulation <strong>of</strong> an anti-apoptotic and a DNA replication gene.<br />

Enhanced cell survival and proliferation: Numerous genes coding for regulators <strong>of</strong> cell<br />

survival signalling pathways, e.g. the IGF-PI3K-PKB pathway, anti-apoptosis, mitosis,<br />

growth and proliferation (incl. proto-oncogenes) were primarily upregulated in Eker rats,<br />

while a tumor suppressor gene and a gene coding for a signal cascade inhibitor (OVCA2)<br />

was downregulated. In comparison, the latter genes were not or only marginally<br />

deregulated in the wild type rats treated with OTA.<br />

48


Chapter 3: Manuscript I<br />

Cell cycle progression and mitosis: Upregulation <strong>of</strong> genes, more directly involved in the<br />

cell cycle progression, was categorized separately as they appeared as an entity distinct<br />

from the category “enhanced cell survival and proliferation”. Remarkably, gene deregulation<br />

that would further cell cycle progression was exclusively observed in Eker rats.<br />

Cell structure remodeling: Similar to the effects observed for genes involved in enhanced<br />

cell survival and proliferation (see above), genes coding for components and regulators <strong>of</strong><br />

the cytoskeleton, altered cell cell-adhesion or communication, and components <strong>of</strong> the Rac<br />

and Rho signalling were almost exclusively upregulated in Eker rats.<br />

Epithelial mesenchymal transition (EMT)/ Fibrosis: This category includes regulators <strong>of</strong><br />

cell proliferation, growth factor activation, cell adhesion and extracellular matrix (ECM)<br />

known to be associated in the process <strong>of</strong> EMT and/ or fibrosis, and which could be<br />

associated with the progression <strong>of</strong> renal and urothelial tumors. Such increased expression<br />

<strong>of</strong> components <strong>of</strong> the TGFß pathway and <strong>of</strong> a hepatocyte growth factor activator-inhibitor<br />

gene in conjunction with a downregulated expression <strong>of</strong> extracellular matrix protease genes<br />

were predominantly deregulated in Eker rats.<br />

49


Chapter 3: Manuscript I<br />

Table 3.2: Categories <strong>of</strong> genes differentially deregulated by OTA in Eker- and wild type rats. For the major pathophysiological categories the associated<br />

genes are listed together with their Genebank accession number. The main biochemical functions or pathways in which these genes are<br />

involved are indicated in column four. For Eker- and wild type rats, the fold deregulation ratios <strong>of</strong> genes that were significantly deregulated<br />

over all time points according to N-way ANOVA are shown in the last eight columns. Genes meeting the significance criteria are indicated in<br />

bold.<br />

Toxicological<br />

category<br />

Metabolism and<br />

Biotransformation<br />

DNA damage<br />

response<br />

Accession<br />

number<br />

Gene title Biochemical category Fold deregulation (mean <strong>of</strong> three replicate animals<br />

per time-point)<br />

NM_031565 CES1B Carboxylesterase RL1 Biotransformation phase I 1.0 -1.7 -2.7 -1.9 -1.1 -1.5 -2.2 -2.2<br />

L46791 CES3 Carboxylesterase 3 Biotransformation phase I 1.0 -1.4 -2.7 -3.4 -1.2 -1.7 -2.7 -1.9<br />

NM_020538 AADAC Arylacetamide deacetylase Biotransformation phase I -1.4 -1.3 -1.9 -2.5 1.0 -1.2 -1.5 -2.0<br />

AW142784 CYP4A12 Cytochrome P450 4a12 Biotransformation phase I 1.0 1.6 1.5 1.9 1.7 1.8 2.0 1.9<br />

NM_133558 CML1 Camello-like 1 Biotransformation phase II -1.1 -1.3 -2.6 -2.9 -1.2 -1.1 -2.0 -2.1<br />

NM_022635 NAT8 N-acetyltransferase 8 Biotransformation phase II -1.2 -2.1 -3.3 -2.9 -1.3 -1.5 -2.4 -2.9<br />

AI072042 GGT6 Gamma-glutamyl transpeptidase type VI Biotransformation phase II -1.4 -2.0 -2.9 -2.7 -1.3 -1.3 -1.6 -1.8<br />

NM_134349 MGST1 Microsomal glutathione S-transferase 1 Biotransformation phase II -1.3 -1.4 -1.8 -1.8 -1.1 -1.3 -1.7 -1.7<br />

NM_134369 CYP2T1 Cytochrome P450 2T1 Biotransformation phase I 1.0 -1.1 -2.0 -1.9 1.1 -2.0 -1.3 -1.3<br />

BF283000 WBSCR21 Williams-Beuren syndr. chromosome region 21 Biotransformation phase I 1.1 -1.4 -1.7 -1.9 -1.1 -1.1 -1.3 -1.4<br />

AI407458 ALDH6A1 Aldehyde dehydrogenase 6A1 Biotransformation phase I 1.8 2.0 1.6 2.1 1.1 1.1 -1.1 -1.1<br />

NM_022270 OCTN1 Organic cation carnitine transporter 1 Drug transport -1.1 -1.1 -2.1 -1.8 1.0 -1.1 -1.5 -1.2<br />

NM_017224 OAT1 Organic anion transporter 1 (SLC22A6) Drug transport -1.1 1.1 -1.9 -1.7 1.1 -1.2 -1.3 -1.6<br />

NM_019303 CYP2F4 Cytochrome P450 2F4 Biotransformation phase I 1.3 1.0 -1.4 -1.8 1.2 -1.8 -1.8 -1.1<br />

NM_017084 GNMT Glycine-N-methyltransferase Biotransformation phase II 1.0 -1.4 -1.4 -1.6 -1.4 -1.5 -1.9 -1.5<br />

U76379 OCT1 Organic cation transporter 1 (SLC22A1) Drug transport 1.1 -1.3 -1.4 -1.7 -1.1 -1.4 -1.7 -1.7<br />

BM388545 SUPT16H Suppressor <strong>of</strong> Ty 16 homolog (predicted) DNA damage repair 1.0 1.8 1.4 1.8 1.6 1.8 2.1 2.4<br />

NM_053677 CHEK2 Checkpoint kinase 2 Cell cycle checkpoint 1.0 1.8 2.8 2.2 1.0 -1.1 1.1 -1.1<br />

Ek<br />

d1<br />

Ek<br />

d3<br />

Ek<br />

d7<br />

Ek<br />

d14<br />

wt<br />

d1<br />

wt<br />

d3<br />

wt<br />

d7<br />

wt<br />

d14<br />

50


Oxidative<br />

stress<br />

response<br />

Enhanced<br />

oxidative stress<br />

Cellular stress<br />

Reduced cell<br />

survival/<br />

proliferation<br />

Accession<br />

number<br />

Chapter 3: Manuscript I<br />

Gene title Biochemical category Fold deregulation (mean <strong>of</strong> three replicate animals<br />

per time-point)<br />

BF548539 MDM2 Ubiquitin E3 ligase Mdm2 (predicted) Cell cycle checkpoint 5.3 4.4 1.8 2.4 -1.4 -1.4 -1.4 1.0<br />

AI178158 RBBP6 retinoblastoma binding protein 6 (predicted) Cell cycle checkpoint 1.0 1.1 1.0 1.0 1.2 1.5 2.0 1.5<br />

NM_019192 SEPP1 Selenoprotein P Oxidative stress response 1.1 3.5 3.4 3.9 4.1 4.2 4.9 4.2<br />

NM_019235 GGTLA1 Gamma-glutamyltransferase-like activity 1 Oxidative stress response 1.4 2.6 1.8 4.2 1.3 -1.4 2.1 1.0<br />

NM_031614 TXNRD1 Thioredoxin reductase 1, cytoplasmic Oxidative stress response 1.7 1.7 1.4 1.7 -1.1 1.0 -1.3 -1.1<br />

AI231438 CN1 Carnosine dipeptidase 1 Oxidative stress response -1.2 -1.2 -2.9 -2.5 -1.2 -1.5 -1.8 -1.3<br />

BM386741 HSP40-3 Heat shock protein hsp40-3 (predicted) Protein folding (cytosol) -1.3 -1.8 -1.7 -1.4 1.0 1.0 -1.5 -1.1<br />

BF414210 KEAP1 Kelch-like ECH-associated protein 1 Regulation <strong>of</strong> transcription 1.7 1.6 2.0 2.4 1.1 1.0 -1.1 1.0<br />

NM_053307 MSRA Methionine sulfoxide reductase A Oxidative stress response 1.4 1.4 1.0 -1.3 1.0 -1.6 -2.0 -1.8<br />

BM387750 DUSP11 Dual specificity phosphatase 11 MAPK pathway -1.1 -1.7 -1.7 -1.5 -1.5 -1.5 -1.4 -1.8<br />

BE110108 DUSP1 Dual specificity protein phosphatase 1 MAPK pathway 1.9 1.6 2.3 2.3 -1.1 -1.3 -1.3 1.0<br />

NM_031032 GMFB Glia maturation factor beta MAPK pathway 2.4 4.0 5.9 5.6 1.5 1.3 2.7 1.5<br />

AAH61870 JNK2 c-Jun N-terminal kinase 2 MAPK pathway 1.8 1.3 1.4 1.3 1.2 -1.4 -1.1 -1.3<br />

AA851481 BRE Brain and reproductive organ-expressed protein MAPK pathway 1.8 1.7 1.5 1.7 1.1 -1.1 -1.2 -1.2<br />

L48060 PRLR Prolactin receptor MAPK pathway 1.5 2.1 1.3 1.7 1.1 1.2 1.2 1.2<br />

NM_023090 HIF2 alpha Hypoxia-inducible factor 2 alpha HIF pathway 2.5 1.8 2.0 1.7 1.2 1.1 -1.2 1.0<br />

BF403837 NFE2L1 Nuclear factor (erythroid-derived) 2 like 1 Regulation <strong>of</strong> gene expression 1.4 1.3 1.3 1.7 1.0 -1.3 -1.1 -1.1<br />

AI598399 RBM3 RNA-binding motif protein 3 RNA metabolism 1.0 1.0 -1.3 1.0 1.4 1.1 1.2 1.8<br />

BF281976 POLD4 Polymerase (DNA-directed), delta 4 DNA replication 2.0 -2.2 -2.0 -2.1 -1.1 -1.4 -1.6 -2.0<br />

NM_057138 CFLAR CASP8 and FADD-like apoptosis regulator Antiapoptotic -1.3 -1.9 -2.9 -4.3 1.2 -3.6 -2.9 -3.4<br />

AW253957 ENDOG Endonuclease G Proapoptotic 1.1 1.9 2.3 1.9 1.3 1.4 1.8 2.5<br />

AA944698 BAT3 HLA-B associated transcript 3 Proapoptotic 6.5 4.4 3.9 5.0 1.2 1.3 1.1 -1.1<br />

NM_019208 MEN1 Multiple endocrine neoplasia 1 Tumor suppressor gene 1.9 1.2 1.6 1.9 -1.1 -1.2 1.1 -1.1<br />

Ek<br />

d1<br />

Ek<br />

d3<br />

Ek<br />

d7<br />

Ek<br />

d14<br />

wt<br />

d1<br />

wt<br />

d3<br />

wt<br />

d7<br />

wt<br />

d14<br />

51


Increased cell<br />

survival/<br />

proliferation<br />

Accession<br />

number<br />

AI113091 TSSC4 Tumor-suppressing subchromosomal transferable<br />

fragment 4 (predicted)<br />

Chapter 3: Manuscript I<br />

Gene title Biochemical category Fold deregulation (mean <strong>of</strong> three replicate animals<br />

per time-point)<br />

Ek<br />

d1<br />

Ek<br />

d3<br />

Ek<br />

d7<br />

Ek<br />

d14<br />

Tumor suppressor gene 1.8 1.4 1.2 2.2 -1.1 1.0 -1.1 1.0<br />

AA996685 PKIA cAMP-dependent protein kinase inhibitor alpha Signalling cascades 3.0 3.1 2.5 2.4 -1.3 1.2 1.3 2.6<br />

BI290885 FSTL Follistatin-like 1 Signalling cascades 1.9 1.2 1.7 1.9 1.1 -1.1 1.1 1.3<br />

BG663107 AKAP12 A kinase anchor protein 12 (gravin) Signalling cascades 1.7 1.4 2.0 1.5 -1.1 -1.2 -1.1 -1.2<br />

M86708 ID1 Inhibitor <strong>of</strong> DNA binding 1 Regulation <strong>of</strong> transcription 1.1 -1.3 1.4 1.0 -1.3 -1.1 -1.3 -1.9<br />

NM_031546 RGN Regucalcin (SMP-30) Antiapoptotic -1.4 1.9 -3.7 -4.5 1.2 -1.7 -1.9 -2.6<br />

BE108969 IGFBP-4 Insulin-like growth factor binding protein 4 (IGF)-PI3K-AKT pathway -1.3 -1.4 -3.0 -3.8 1.0 -1.3 -3.0 -2.8<br />

AI145815 MKRN-1 Makorin-1 Protein metabolism -1.1 -1.6 -1.9 -1.9 -1.3 -1.7 -1.7 -1.6<br />

BM986536 HIST1H4I Histone 1 H4i Nucleosome assembly 1.4 3.1 2.7 3.0 1.3 2.2 2.5 2.3<br />

NM_022265 PDC4 Programmed cell death 4 Antiapoptotic 1.5 2.1 1.7 1.9 1.0 -1.1 -1.3 1.1<br />

BE112895 PEA15 Phosphoprotein enriched in astrocytes 15 Antiapoptotic -1.5 1.3 1.9 1.9 1.2 1.1 1.4 1.1<br />

NM_021846 MCL1 Myeloid cell leukemia 1 Antiapoptotic 1.6 1.4 1.5 1.8 1.1 -1.3 -1.1 -1.4<br />

NM_022943 MERTK C-Mer proto-oncogene tyrosine kinase Protooncogene 3.3 4.9 4.3 2.9 -1.3 1.0 1.2 1.1<br />

NM_012807 SMOH Smoothened Protooncogene 1.8 1.3 1.6 1.5 -1.1 1.0 -1.3 1.0<br />

NM_022264 V-KIT Hardy-Zuckerman 4 feline sarcoma viral oncogene<br />

homolog (c-Kit)<br />

NM_012843 TMP Tumor-associated membrane protein (EMP1 Epithelial<br />

membrane protein 1)<br />

Protooncogene 2.9 3.7 2.4 2.5 -1.2 -1.3 -1.5 1.2<br />

Protooncogene 2.8 1.7 2.2 4.3 1.0 -1.3 -1.2 -1.3<br />

AA943541 OVCA2 Candidate tumor suppressor OVCA2 (predicted) Tumor suppressor gene -1.3 -1.4 -1.6 -1.8 -1.3 -1.1 -1.4 -1.6<br />

NM_053481 PIK3CB Phosphatidylinositol-4,5-bisphosphate 3-kinase<br />

catalytic beta subunit<br />

(IGF)-PI3K-AKT pathway 2.0 2.9 -1.3 2.0 1.3 1.0 1.0 -1.1<br />

AI102030 AKT1S1 AKT1 substrate 1 (proline-rich) (IGF)-PI3K-AKT pathway 2.4 4.0 2.0 2.1 1.0 1.0 -1.1 -1.1<br />

AI105076 AKT2 Thymoma viral proto-oncogene 2 (IGF)-PI3K-AKT pathway 1.5 1.5 1.2 1.9 1.0 1.1 -1.1 -1.2<br />

AW434982 SBF1 Similar to SET binding factor 1 (IGF)-PI3K-AKT pathway 2.4 3.5 2.2 3.6 1.2 1.0 -1.1 1.0<br />

wt<br />

d1<br />

wt<br />

d3<br />

wt<br />

d7<br />

wt<br />

d14<br />

52


Cell cycle<br />

progression<br />

and mitosis<br />

Accession<br />

number<br />

Chapter 3: Manuscript I<br />

Gene title Biochemical category Fold deregulation (mean <strong>of</strong> three replicate animals<br />

per time-point)<br />

NM_012593 KLK7 Glandular kallikrein 7 Protein metabolism 1.8 1.3 1.9 1.6 -1.1 -1.2 -1.2 -1.1<br />

BI300565 ADAM10 A-disintegrin and metalloprotease domain 10 Protein metabolism 1.6 1.8 1.3 1.4 1.1 -1.3 -1.3 -1.3<br />

NM_053665 AKAP1 A kinase (PRKA) anchor protein 1 Signalling cascades 5.9 3.8 4.1 7.8 1.0 1.2 -1.1 1.0<br />

NM_017094 GHR Growth hormone receptor Signalling cascades 1.7 2.8 1.2 2.0 1.2 -1.1 1.0 1.1<br />

NM_012850 GHRHR Growth hormone releasing hormone receptor Signalling cascades 1.2 1.3 1.8 1.7 1.6 1.4 1.1 -1.1<br />

BI294916 KLF2 Kruppel-like factor 2 Regulation <strong>of</strong> gene expression 1.6 1.4 1.9 1.9 1.1 -1.1 -1.2 1.2<br />

NM_131904 MGEA5 Meningioma expressed antigen 5 Protein metabolism 2.1 1.7 1.8 1.4 -1.1 -1.1 -1.5 -1.7<br />

AF080594 VEGF Vascular endothelial growth factor Angiogenesis 2.3 2.0 1.6 2.6 1.1 -1.3 1.0 1.0<br />

AW524517 VEZF1 Vascular endothelial zinc finger 1 Angiogenesis 1.9 1.6 1.2 1.4 1.1 1.0 -1.1 1.1<br />

NM_133569 ANGPTL2 Angiopoietin-like 2 Angiogenesis 1.3 1.9 1.2 1.9 1.2 -1.1 -1.1 1.1<br />

NM_017089 EPHB1 Ephrin B1 Angiogenesis 1.9 1.6 1.6 1.9 1.1 1.0 -1.3 1.0<br />

AB035507 MCAM Melanoma cell adhesion molecule Angiogenesis 2.1 1.5 1.6 1.3 1.0 -1.3 1.1 -1.1<br />

NM_022615 TOP1 DNA topoisomerase 1 DNA replication 1.5 1.8 1.6 2.2 1.0 -1.1 1.0 1.1<br />

BM385181 SMARCA4 SWI-SNF related matrix associated actin<br />

dependent regulator <strong>of</strong> chromatin subfamily A4<br />

Ek<br />

d1<br />

Ek<br />

d3<br />

Ek<br />

d7<br />

Ek<br />

d14<br />

Chromatin remodelling 1.3 1.1 1.7 1.7 -1.1 1.4 1.0 1.1<br />

U75920 MAPRE1 Microtubule-associated protein RP/EB family 1 Mitotic spindle formation 2.4 2.6 2.0 2.8 1.1 1.0 -1.1 1.0<br />

AJ306292 AJUBA Ajuba protein Mitotic spindle formation 1.4 1.3 1.3 1.8 1.0 -1.2 -1.1 -1.2<br />

BE118382 NEK9 NIMA related kinase 9 Mitotic spindle formation 2.1 1.9 1.8 2.4 1.0 1.0 1.0 -1.1<br />

U77583 CSNK1A1 Casein kinase 1, alpha 1 Mitotic spindle formation 1.6 1.5 1.6 1.8 1.1 1.0 -1.1 1.0<br />

NM_057148 SEPT2 Septin 2 Mitotic spindle formation 2.5 2.2 2.1 2.5 -1.1 -1.1 1.0 -1.1<br />

NM_080907 PPP4R1 Protein phosphatase 4, regulatory subunit 1 Mitotic spindle formation 2.1 1.7 1.3 2.0 1.0 1.0 -1.2 -1.1<br />

NM_013194 NMMHC-A Nonmuscle myosin heavy chain A Mitotic spindle formation 1.6 1.6 1.7 2.0 1.3 -1.1 1.1 1.2<br />

Cell structure AA955773 PCDH1 Protocadherin 1 Cell adhesion molecule -1.1 -1.7 -1.6 -1.9 -1.3 -1.7 -2.2 -1.8<br />

wt<br />

d1<br />

wt<br />

d3<br />

wt<br />

d7<br />

wt<br />

d14<br />

53


emodelling<br />

Accession<br />

number<br />

Chapter 3: Manuscript I<br />

Gene title Biochemical category Fold deregulation (mean <strong>of</strong> three replicate animals<br />

per time-point)<br />

BE097805 PCDH4 Protocadherin 4 Cell adhesion molecule 2.7 1.2 3.6 2.3 1.5 1.3 1.6 1.3<br />

AA955091 ITGA6 Integrin alpha 6 Cell adhesion molecule 2.0 1.3 1.7 1.5 1.0 1.0 -1.3 -1.4<br />

NM_031699 CLDN1 Claudin 1 Cell adhesion molecule 1.6 1.2 2.0 1.8 1.0 -1.2 -1.3 -1.2<br />

NM_031329 OCLN Occludin Cell adhesion molecule 2.5 1.9 1.3 2.2 1.4 1.0 -1.1 1.0<br />

NM_013217 AF-6 Afadin Regulation <strong>of</strong> cell adhesion/<br />

migration<br />

BF284125 IQGAP1 IQ motif containing GTPase activating protein 1 Regulation <strong>of</strong> cell adhesion/<br />

migration<br />

AB020726 PODXL Podocalyxin Regulation <strong>of</strong> cell adhesion/<br />

migration<br />

NM_020085 RPTPK Receptor-like protein tyrosine phosphatase kappa<br />

extracellular region<br />

Ek<br />

d1<br />

Ek<br />

d3<br />

Ek<br />

d7<br />

Ek<br />

d14<br />

wt<br />

d1<br />

wt<br />

d3<br />

wt<br />

d7<br />

wt<br />

d14<br />

1.6 1.7 1.7 1.9 1.0 1.0 -1.2 -1.2<br />

1.3 1.3 1.8 1.8 1.0 -1.1 -1.3 -1.2<br />

1.6 1.3 1.5 1.8 1.2 1.0 1.1 -1.1<br />

Signalling cascades 5.6 5.7 3.2 5.4 1.0 -1.4 -1.8 -1.4<br />

NM_057115 PTPN12 Protein tyrosine phosphatase, non-receptor type12 Signalling cascades 1.9 1.5 2.7 3.6 -1.1 -1.3 -1.2 -1.2<br />

NM_031034 GNA12 Guanine nucleotide-binding protein alpha 12 Signalling cascades 2.2 2.2 1.6 2.4 1.1 1.2 1.1 -1.1<br />

BE115857 PARVA Parvin alpha Cytoskeleton organization 2.4 2.1 1.6 2.1 1.0 1.0 1.1 -1.1<br />

NM_024401 AVIL Advillin (Pervin) Cytoskeleton organization 1.2 1.3 1.9 2.6 1.0 1.1 1.5 1.8<br />

AF054618 CTTN Cortactin Cytoskeleton organization 1.8 1.5 1.9 2.0 1.1 1.0 1.1 1.1<br />

NM_032613 LASP1 LIM and SH3 protein 1 Cytoskeleton organization 3.1 1.8 1.5 2.0 1.1 1.0 -1.2 1.1<br />

NM_023982 ARHGEF11 Rho guanine nucleotide exchange factor 11 Cytoskeleton organization 1.7 2.5 2.8 3.1 1.7 2.1 2.1 1.4<br />

AI170442 DSTN Destrin Cytoskeleton organization 1.5 1.5 1.7 2.0 1.0 1.1 1.0 -1.1<br />

NM_030873 PFN2 Pr<strong>of</strong>ilin 2 Cytoskeleton organization 1.6 2.5 2.1 3.4 1.3 -1.1 -1.2 1.2<br />

AA875047 CCTZ Chaperonin containing T-complex 1 zeta Cytoskeleton organization 9.0 2.0 6.7 5.0 -2.2 2.4 -1.5 -1.1<br />

V01217 ACTB Actin beta Cytoskeleton constituent 2.2 2.1 2.1 3.1 1.0 -1.1 -1.1 1.0<br />

NM_016990 ADD1 Alpha adducin Cytoskeleton constituent 2.0 1.3 1.5 2.1 1.2 1.2 -1.1 1.1<br />

X70706 PLS3 Plastin 3 T-is<strong>of</strong>orm (T-plastin) Cytoskeleton constituent 2.0 2.4 1.8 2.9 1.0 1.1 1.0 1.2<br />

54


EMT / Fibrosis<br />

Accession<br />

number<br />

Chapter 3: Manuscript I<br />

Gene title Biochemical category Fold deregulation (mean <strong>of</strong> three replicate animals<br />

per time-point)<br />

NM_030863 MSN Moesin Cytoskeleton constituent 1.7 1.8 1.6 1.9 1.3 -1.2 1.0 1.1<br />

BF392456 SPNB2 Spectrin beta 2 Cytoskeleton constituent 1.9 1.9 1.5 2.4 1.1 1.1 1.0 1.0<br />

BI284344 KRT2-7 Keratin complex 2, basic, gene 7 (predicted) Cytoskeleton constituent -1.1 -1.4 -1.8 -2.0 1.0 -1.3 -1.4 -1.6<br />

BI295970 TPM3 Tropomyosin 3 gamma (Tropomyosin alpha 3) Cell morphology / motility 1.7 1.7 1.6 2.1 1.0 1.0 -1.1 -1.1<br />

NM_012678 TPM4 Tropomyosin 4 Cell morphology / motility 2.2 1.8 2.3 2.0 1.0 -1.1 -1.4 1.1<br />

NM_053986 MYO1B Myosin IB (Brush border myosin I) Cell morphology / motility 1.7 1.3 1.5 1.8 -1.1 1.1 -1.1 -1.1<br />

NM_023092 MYO1C Myosin 1C (Unconventional myosin Myr2 I heavy<br />

chain)<br />

Ek<br />

d1<br />

Ek<br />

d3<br />

Ek<br />

d7<br />

Ek<br />

d14<br />

Cell morphology / motility 1.5 1.4 1.7 1.7 1.1 1.1 1.0 1.1<br />

BG380723 CAP1 Adenylyl cyclase-associated protein 1 Cell morphology / motility 2.0 1.5 1.9 2.3 -1.1 1.2 1.0 1.0<br />

AA997129 LAMC1 Laminin gamma 1 Extracellular matrix component 2.2 1.5 1.6 1.6 1.1 -1.2 1.0 1.3<br />

AW435213 NANS N-acetylneuraminic acid synthase (sialic acid<br />

synthase)<br />

Cell adhesion / migration 1.4 1.3 1.5 1.7 1.0 1.1 -1.3 -1.2<br />

U61261 LAMA3 Laminin alpha 3 (Laminin-5 alpha 3) Extracellular matrix component 1.2 1.1 1.2 -1.3 -1.2 -1.7 -1.6 -2.0<br />

BF284673 FGD1 Faciogenital dysplasia homolog Cell adhesion/ migration 1.1 1.3 1.2 1.5 1.5 1.2 1.9 2.2<br />

NM_013085 PLAU Urokinase-type plasminogen activator Cell adhesion/ migration -1.3 -1.3 -2.1 -1.9 -1.3 -2.0 -1.7 -2.1<br />

NM_013143 MEP1A Meprin A alpha-subunit Protein degradation -1.2 1.0 -2.5 -2.9 -1.2 -1.6 -1.6 -2.1<br />

L09653 TGFBR2 TGF-beta receptor type II TGF-beta family pathway 3.9 5.5 5.9 3.4 1.0 -1.4 -1.1 1.0<br />

NM_013130 SMAD1 (MADH1) Mothers against decapentaplegic homolog<br />

1<br />

NM_013095 SMAD3 (MADH3) Mothers against decapentaplegic homolog<br />

3<br />

NM_019275 SMAD4 (MADH4) Mothers against decapentaplegic homolog<br />

4)<br />

TGF-beta family pathway 2.1 1.5 1.6 1.4 1.0 1.1 1.0 1.7<br />

TGF-beta family pathway 2.1 3.5 1.6 2.8 1.2 1.1 -1.1 1.1<br />

TGF-beta family pathway 1.7 1.7 1.3 1.6 -1.2 -1.2 -1.3 -1.6<br />

NM_053357 CTNNB1 Beta-catenin Regulation <strong>of</strong> proliferation 1.7 1.6 1.3 1.9 1.0 -1.2 -1.1 1.0<br />

NM_031317 PDGFC Platelet derived growth factor C Regulation <strong>of</strong> proliferation 2.1 1.7 1.4 2.4 1.0 -1.4 -1.1 -1.1<br />

wt<br />

d1<br />

wt<br />

d3<br />

wt<br />

d7<br />

wt<br />

d14<br />

55


Accession<br />

number<br />

Chapter 3: Manuscript I<br />

Gene title Biochemical category Fold deregulation (mean <strong>of</strong> three replicate animals<br />

per time-point)<br />

BE095528 HAI-1 Hepatocyte growth factor activator inhibitor 1 Regulation <strong>of</strong> proliferation 3.9 9.2 5.4 3.0 1.0 1.0 -1.1 1.0<br />

AF172255 NPHS1 Nephrosis 1 homolog (nephrin) Cell adhesion / migration 1.7 2.3 2.0 1.8 -1.1 1.0 1.0 1.1<br />

NM_012886 TIMP3 Tissue inhibitor <strong>of</strong> metalloproteinase 3 Cell adhesion / migration 1.8 1.4 1.4 1.8 1.0 -1.1 -1.4 -1.7<br />

BI278545 DPT Dermatopontin Cell adhesion / migration 1.6 1.5 1.7 1.8 1.1 1.0 1.2 1.2<br />

NM_012924 CD44 antigen (ECMR-III Extracellular matrix receptor-III) Cell adhesion / migration 2.3 3.3 1.2 1.6 -1.2 -1.1 1.1 1.2<br />

NM_024358 NOTCH2 Notch homolg 2 Differentiation / organogenesis 2.4 2.2 3.2 7.4 1.0 1.0 -1.1 -1.1<br />

AA997458 Similar to CSRP2 Cysteine and glycine-rich protein 2 Differentiation / organogenesis 1.0 -1.5 -1.8 -1.6 -1.3 -1.5 -1.6 -1.6<br />

NM_012901 AMBP Alpha-1-microglobulin/bikunin precursor Extracellular transport 1.1 2.0 2.6 1.9 1.0 1.3 1.8 1.9<br />

NM_017309 CNB Calcineurin B regulatory subunit is<strong>of</strong>orm 1 Signalling cascades 4.5 7.6 1.8 5.8 -1.1 -1.4 -1.3 -1.3<br />

Ek<br />

d1<br />

Ek<br />

d3<br />

Ek<br />

d7<br />

Ek<br />

d14<br />

wt<br />

d1<br />

wt<br />

d3<br />

wt<br />

d7<br />

wt<br />

d14<br />

56


3.5 Discussion<br />

Chapter 3: Manuscript I<br />

As expected for the short-term duration and dose-regimen employed (Mengs et al. 1982;<br />

Boorman et al. 1992), neither AA nor OTA induced pronounced non-neoplastic renal<br />

pathology in either strain <strong>of</strong> rats. While AA treated rats presented with a slightly higher<br />

inflammatory response than the corresponding controls, OTA treated rats responded with<br />

the typical pathological changes, e.g. karyomegaly, apoptosis, cell shedding, regenerative<br />

proliferation and an inflammatory response in the renal cortex, as reported earlier (Boorman<br />

et al. 1992; Rasonyi et al. 1999). However, the AA and OTA induced non-neoplastic<br />

pathology was clearly distinct from one another, and this was also reflected in the cell<br />

proliferation assessment (Figure 3.1). Lack <strong>of</strong> overt cell necrosis and subsequent cell<br />

shedding and regeneration in the tubuli <strong>of</strong> AA treated rats coincided with absence <strong>of</strong><br />

increased cell proliferation, demonstrating that AA had neither a cytotoxic nor a mitogenic<br />

effect on the kidneys <strong>of</strong> either strain <strong>of</strong> rats. In contrast, OTA- induced tubular toxicity and<br />

possibly mitogenic activity was reflected in the overt cell regeneration observed with the<br />

histopathological as well as immunohistological assessments. Moreover, the observed<br />

effects increased with the duration <strong>of</strong> treatment and were comparable in extent <strong>of</strong> effect in<br />

both strains. Based on above observations, the age <strong>of</strong> animals used and the duration and<br />

type <strong>of</strong> exposure, the expectation was confirmed that no increased prevalence (no. <strong>of</strong><br />

animals affected) or number (no. <strong>of</strong> lesions per animal) <strong>of</strong> preneoplastic and neoplastic<br />

lesions were to be encountered in the wild type control and AA treated rats. Moreover, AA<br />

treatment <strong>of</strong> the arguably more susceptible Eker rats did not increase the prevalence or<br />

number <strong>of</strong> lesions, suggesting that the Tsc2 mutation was not critical for AA induced<br />

effects. The two carcinomas observed on day 14 in AA exposed Eker rats cannot be<br />

conclusively associated with AA treatment, as adenomas were also observed in the<br />

corresponding Eker rat control and AA treatment groups already on day 1 (Supplemetary<br />

table 3.2). As the number <strong>of</strong> animals employed for this experiment limited statistical<br />

evaluation, a progression <strong>of</strong> already present adenomas to carcinomas due to AA treatment<br />

cannot be excluded.<br />

Similar to AA, OTA treatment was not associated with an increased prevalence or number<br />

<strong>of</strong> lesions in wild type rats, despite the clearly enhanced cell proliferative response in the<br />

renal cortex. The latter observation corroborates numerous earlier findings (Boorman et al.<br />

1992; Rasonyi et al. 1999) suggesting that non-genotoxic compound- induced renal<br />

carcinogenesis can only be observed after a prolonged compound exposure period. In<br />

contrast to the situation in wild type, Eker rats presented with a significantly increased<br />

57


Chapter 3: Manuscript I<br />

prevalence and number <strong>of</strong> AT on day 14 <strong>of</strong> OTA treatment (Figure 3.1, E, Supplementary<br />

table 3.2). The latter finding suggests that the increased cell proliferative stimulus (Figure<br />

3.1, C) provided for an enhanced manifestation <strong>of</strong> the predisposition for renal neoplasia<br />

mediated by the Tsc2 mutation and thus a direct or indirect interaction <strong>of</strong> OTA with Tsc2<br />

(tuberin) pathway.<br />

At the outset <strong>of</strong> this experiment it was assumed that each compound would induce a<br />

distinct gene expression pr<strong>of</strong>ile, which is reflected by the short-term pathology but also<br />

displays characteristic genes representative for pathways involved in the compoundspecific<br />

type <strong>of</strong> renal carcinogenesis. Consequently, the genotoxic AA was expected to<br />

induce a gene expression pr<strong>of</strong>ile, most likely involving genes <strong>of</strong> cell cycle arrest and DNA<br />

damage repair but not cell proliferation and most likely would not involve Tsc2. Indeed, the<br />

expression pr<strong>of</strong>iles <strong>of</strong> AA- treated Eker and wild type rats (Figure 3.3) were not distinctly<br />

different and were comparable to expression pr<strong>of</strong>iles obtained with AA in kidneys <strong>of</strong> Big<br />

Blue transgenic F344 rats (Chen et al. 2006). Several phase I genes were deregulated by<br />

AA treatment from the first day <strong>of</strong> exposure. The gene product <strong>of</strong> one <strong>of</strong> the upregulated<br />

phase I genes, NQOI, was previously demonstrated to be capable <strong>of</strong> reducing the nitro<br />

group <strong>of</strong> AAI leading to metabolic activation. NQOI up-regulation could therefore be at least<br />

partly responsible for DNA adduct formation (Stiborova et al. 2003). Although DNA adduct<br />

formation could not be measured with the study design employed, previously published<br />

reports support the assumption that AA DNA-adducts are formed (Dong et al. 2006).<br />

Indeed, upregulation <strong>of</strong> several p53 pathway genes, including p53 target genes carrying a<br />

p53 consensus sequence in the promotor region, was most prominent on day 7 and 14 <strong>of</strong><br />

exposure, indicating a DNA damage response upon bioactivation <strong>of</strong> AA. This interpretation<br />

is further supported by the fact that several <strong>of</strong> the p53 target genes, e.g. MDM2, p21 or<br />

CCNG1, have also been shown to be upregulated in rat liver after short-term exposure to<br />

different known genotoxic compounds (Ellinger-Ziegelbauer et al. 2005). DNA damage and<br />

activation <strong>of</strong> p53 pathway genes are expected to result in cell cycle arrest (Figure 3.4, A),<br />

followed by damage repair or programmed cell death (Sionov and Haupt 1999). AAtreatment<br />

led to the deregulation <strong>of</strong> several genes involved in apoptosis, with a comparable<br />

time pr<strong>of</strong>ile to the p53- pathway and target genes. Although cell cycle components are<br />

predominantly regulated on the protein level, downregulation <strong>of</strong> genes crucial for the G2/M<br />

transition, e.g. CDC2, Cyclin-B, TOME1 and CKS2 and downregulation <strong>of</strong> genes required<br />

for mitotic spindle formation like TUBA1 or HMMR, suggest a G2 arrest. Deregulation <strong>of</strong> the<br />

latter genes in wild type rats only, may be explained by 12-fold lower PI3K mRNA<br />

expression compared to Eker rats (Sen et al. 2004). Increasing evidence suggest that<br />

constitutive activation <strong>of</strong> the PI3K pathway could lead to defects in DNA damage checkpoint<br />

control (Liang and Slingerland 2003). Consequently, AA-induced G2/M arrest would<br />

58


Chapter 3: Manuscript I<br />

therefore not be readily detectable in Eker rats, although they responded with upregulation<br />

<strong>of</strong> proapoptotic genes, as well as downregulation <strong>of</strong> Ki-67, an observation also supported<br />

by the cell proliferation and pathological analysis (Figure 3.1, A and B).<br />

Figure 3.4: Postulated mechanistic pathways <strong>of</strong> AA (A) and OTA (B) induced toxicity in Eker and<br />

wild type rats and potential influence <strong>of</strong> Tsc2 in the manifestation <strong>of</strong> short- and longterm<br />

effects discussed. Black: pathways and processes suggested from the literature;<br />

Blue: pathways and processes implicated by gene expression analysis; Red:<br />

Pathways and processes implicated by histopathology and corroborated by gene<br />

expression analysis.<br />

In contrast to AA, the gene expression pr<strong>of</strong>iles <strong>of</strong> OTA-treated Eker and wild type rats were<br />

distinctly different (Figure 3.3). However, in both strains up-regulation <strong>of</strong> CYP4A12 and<br />

down-regulation <strong>of</strong> other phase I and II genes could provide for increased generation <strong>of</strong><br />

reactive oxygen species (ROS) and enhanced oxidative DNA damage (Kamp et al. 2005;<br />

Mally et al. 2005), that could lead to cellular damage and regenerative cell proliferation.<br />

Indeed, enhanced proximal tubular damage and cell regeneration/proliferation (Figure 3.1,<br />

C and D) was observed in both rat strains, corroborating the above interpretation. OTA<br />

induced oxidative stress and ensuing DNA damage in combination with enhanced cell<br />

59


Chapter 3: Manuscript I<br />

proliferation could increase the likelihood <strong>of</strong> neoplastic transformation (Dietrich and<br />

Swenberg 1991). Presence <strong>of</strong> oxidative DNA damage, as also suggested by earlier findings<br />

(Luhe et al. 2003; Cavin et al. 2006; Marin-Kuan et al. 2006), is supported by the<br />

upregulation <strong>of</strong> the p53 pathway genes SUPT16H in both strains, MDM2 and CHEK2 in<br />

Eker rats, and RBBP6 in wild type rats, as well as by the time-dependent downregulation <strong>of</strong><br />

HSP 40-3, CN1, MSRA and MGST1, responsible for the protection <strong>of</strong> cells against oxidative<br />

stress. Indeed, recent findings demonstrated that overexpression <strong>of</strong> glia maturation growth<br />

factor beta (GMFB) resulted in reduced antioxidant enzyme activities, subsequent<br />

accumulation <strong>of</strong> H2O2, and finally enhanced oxidative injury <strong>of</strong> renal proximal tubular cells<br />

(Kaimori et al. 2003). The 5.9-fold upregulation <strong>of</strong> GMFB in Eker rats, shown here, and the<br />

known reduced 8-oxoguanine-DNA glycosylase expression in Eker rats (Habib et al. 2003),<br />

may suggest that Eker rats are more susceptible to oxidative stress than wild type rats. This<br />

interpretation is supported by the observation that Eker rats responded with increased cell<br />

proliferation already on day 7 (Figure 3.1, C) and increased formation <strong>of</strong> AT (Figure 3.1, E).<br />

However, OTA induced regenerative proliferation may not have been the sole contributor to<br />

the propagation <strong>of</strong> preneoplastic lesions. Indeed, OTA has previously been assumed to<br />

have mitogenic properties (Horvath et al. 2002; Gekle et al. 2005), a hypothesis supported<br />

by downregulation <strong>of</strong> IGFBP-4, a negative regulator <strong>of</strong> the (IGF)-PI3K-AKT mitogenic<br />

pathway, in Eker and wild type rats. Despite this, only Eker rats demonstrated a significant<br />

increase <strong>of</strong> preneoplastic lesions. The latter can be explained with the renal gene<br />

expression pr<strong>of</strong>ile <strong>of</strong> OTA-treated Eker rats which contains deregulated genes characteristic<br />

<strong>of</strong> the most pertinent hallmarks involved in cancer progression (Hanahan and Weinberg<br />

2000), namely: 1) the evasion <strong>of</strong> programmed cell death via upregulation <strong>of</strong> antiapoptotic<br />

genes e.g. PDC 4, PEA15 or MCL1; 2) insensitivity to growth inhibitory signals via<br />

upregulation <strong>of</strong> protooncogenes e.g. MERTK, SMOH, V-KIT, TMP; 3) self sufficiency in<br />

growth signals e.g. via an activated (IGF)-PI3K-AKT pathway (PIK3CB, AKT1S1, AKT2 or<br />

SBF1); 4) limitless replication potential e.g. via an upregulation <strong>of</strong> genes involved in DNA<br />

replication (TOP1), chromatin remodelling (SMARCA4), or mitotic spindle formation<br />

(MAPRE1, AJUBA, NEK9); 5) sustained angiogenesis via upregulation <strong>of</strong> VEGF, VEZF1,<br />

ANGPTL2; and 6) tissue invasion and metastasis via a broad set <strong>of</strong> deregulated genes<br />

involved in cell structure remodelling (e.g. Rho guanine nucleotide exchange factor<br />

ARHGEF11 or IQGAP1, an effector for CDC42 and RAC1) and EMT (TGFßR-II, SMADs,<br />

CD44, TIMP3).<br />

As demonstrated, OTA treatment <strong>of</strong> Eker rats led to the expression <strong>of</strong> genes that could be<br />

intricately involved in, or are the result <strong>of</strong>, activated mTor signalling (Thomas 2006). Thus<br />

OTA treatment and the Tsc2-mutation may have acted in concert or separately on the<br />

(IGF)-PI3K-AKT pathway thus resulting in a co-joint activation <strong>of</strong> mTor signalling. Additional<br />

60


Chapter 3: Manuscript I<br />

stimulation <strong>of</strong> the mTor pathway could be possible by OTA-mediated ERK activation<br />

(Horvath et al. 2002). Tsc2 was demonstrated to be a direct substrate <strong>of</strong> ERK (Ma et al.<br />

2005a). As Eker rats exhibit only one functional allele <strong>of</strong> the Tsc2 gene, OTA mediated<br />

ERK-dependent inactivation <strong>of</strong> Tsc2 could lead to more drastic and therefore earlier<br />

detection <strong>of</strong> proliferative effects and neoplastic transformation in Eker rats (Figure 3.4, B).<br />

In summary, gene expression pr<strong>of</strong>ile comparisons with histopathologic findings from AAand<br />

OTA- treated Eker and wild type rats discussed here, highlight that gene expression<br />

analysis subsequent to short-term in vivo assays may have the potential to identify<br />

deregulated genes involved in compound- and strain- specific pathology. Moreover,<br />

deregulation <strong>of</strong> genes, for which a similar direction <strong>of</strong> deregulation has been reported for<br />

various types <strong>of</strong> cancers, suggests that pathways linked to tumorigenesis may be<br />

deregulated already after short-term carcinogen exposure. Whether these changes in gene<br />

expression are transient or can be causally linked to a compound-specific tumorigenicity<br />

cannot be determined without gene expression pr<strong>of</strong>ile analysis <strong>of</strong> the respective<br />

preneoplastic and neoplastic lesions in rats chronically treated with AA or OTA. This<br />

analysis, as currently carried out in this laboratory using laser-capture microdissection<br />

(Stemmer et al. 2006) will provide further information as to the relevance <strong>of</strong> the pathways<br />

identified in short-term experiments for the understanding <strong>of</strong> the mechanisms underlying<br />

AA- and OTA- induced renal carcinogenesis.<br />

Acknowledgements<br />

We would like to thank the Federal Ministry <strong>of</strong> Education and Research for funding the<br />

project (BMBF: 0313024), Tanja Lampertsdörfer and Gudrun von Scheven for skillful<br />

assistance during the whole animal experiment, Evelyn O’Brien and Alexandra Heussner<br />

for help with the animal sacrifice, and Margot Thiel and Kerstin Lotz for microarray<br />

hybridization.<br />

61


3.6 Supplement<br />

3.6.1 Supplementary Information<br />

Chapter 3: Manuscript I<br />

The significance <strong>of</strong> microarray data for biological interpretations and the necessity <strong>of</strong><br />

confirming them by another method were extensively discussed by Rockett et al. (Rockett<br />

and Hellmann 2004). The authors suggested that microarrays as well as quantitative realtime<br />

polymerase chain reaction (QRT-PCR) are at best semi-quantitative, while quantitative<br />

detection <strong>of</strong> gene expression cannot be achieved with any <strong>of</strong> the currently available<br />

methodologies. Based on 284 individual comparisons, a regression coefficient <strong>of</strong> 0.94<br />

between the natural log ratios for Affymetrix RAE230A arrays and QRT-PCR (Taqman®<br />

technology) data was previously obtained in this laboratory (data not shown). This supports<br />

the assumption that both technologies deliver at least semi-quantitatively comparable<br />

expression ratios. Therefore, it was assumed that the expression pr<strong>of</strong>iles generated with<br />

the Affymetrix platform, as presented here, can be used for semi-quantitative comparisons<br />

<strong>of</strong> gene deregulation and subsequent data interpretation.<br />

62


3.6.2 Supplementary Tables<br />

Supplementary table 3.1: Numbers and categories <strong>of</strong> genes deregulated by AA or OTA<br />

# genes<br />

Ek<br />

specific<br />

# genes<br />

Ek and wt<br />

specific<br />

# genes<br />

wt<br />

specific<br />

# genes<br />

Ek<br />

specific<br />

# genes<br />

Ek and wt<br />

specific<br />

# genes<br />

wt<br />

specific<br />

Compound AA AA AA OTA OTA OTA<br />

# all significantly<br />

deregulated genes<br />

Pathophysiological<br />

categories<br />

Metabolism and<br />

biotransformation<br />

77 34 47 314 61 80<br />

Description<br />

Chapter 3: Manuscript I<br />

10 12 3 5 6 3 Deregulation <strong>of</strong> genes encoding biotransformation enzymes and drug transporters<br />

DNA damage response 1 7 1 2 1 1 Upregulation <strong>of</strong> genes encoding DNA damage repair enzymes and p53 target genes<br />

Oxidative stress response - - - 2 1 - Upregulation <strong>of</strong> ARE A target genes and other genes known to be involved in oxidative stress<br />

responses<br />

Enhanced oxidative stress - - 1 2 3 1 Downregulation <strong>of</strong> genes mediating the cellular antioxidant defense or upregulation <strong>of</strong> genes<br />

encoding Nrf2 B inhibitors<br />

Response to unfolded and<br />

damaged proteins<br />

1 - - 6 - 2 Upregulation <strong>of</strong> genes encoding members <strong>of</strong> the heat shock- or glucose-regulated protein<br />

family and others involved in the response to unfolded or damaged proteins<br />

Cellular stress 1 - - 6 2 1 Upregulation <strong>of</strong> immediate early genes, target genes <strong>of</strong> the stress kinase-, NFkB C -, or HIF D<br />

pathways or other stress related pathways. Downregulation <strong>of</strong> genes encoding inhibitors <strong>of</strong><br />

these pathways<br />

Inhibited cell survival and<br />

proliferation<br />

Enhanced cell survival/<br />

proliferation<br />

Cell cycle progression and<br />

mitosis<br />

8 1 7 6 3 2 Downregulation <strong>of</strong> antiapoptotic genes, genes involved in cell cycle progression or genes<br />

encoding components <strong>of</strong> the mitotic spindle apparatus. Upregulation <strong>of</strong> proapoptotic genes<br />

or tumour suppressor genes<br />

1 - 3 23 2 - Upregulation <strong>of</strong> genes encoding growth and survival factors, (IGF)- PI3K-AKT E pathway<br />

components, antiapoptotic genes, genes <strong>of</strong> the VEGF F pathway and functioning in growth<br />

and survival <strong>of</strong> endothelial cells. Downregulation <strong>of</strong> tumour suppressor genes.<br />

- - - 10 1 - Upregulation <strong>of</strong> genes involved in DNA replication, mitotic spindle/ cytokineses or<br />

nucleosome formation.<br />

63


Cell structure remodelling<br />

Chapter 3: Manuscript I<br />

2 2 2 32 1 2 Up- and downregulation <strong>of</strong> genes coding for components or regulators <strong>of</strong> the cytoskeleton,<br />

cell adhesion molecules and components <strong>of</strong> the ECM. Upregulation <strong>of</strong> gene transcripts<br />

involved in glycoprotein synthesis.<br />

EMT/ Fibrosis - - - 12 2 - Upregulation <strong>of</strong> genes involved in the TGF-ßG pathway, ECMH accumulation or deregulation<br />

<strong>of</strong> other genes known to be involved in the process <strong>of</strong> epithelial mesenchymal transition<br />

(EMT) and /or fibrosis<br />

Inflammation 8 2 2 13 - 2 Upregulation <strong>of</strong> genes encoding components <strong>of</strong> the TNF I / cytokine pathway, <strong>of</strong> the antigene<br />

presentation machinery, or inflammatory factors. Downregulation <strong>of</strong> inhibitors <strong>of</strong> blood<br />

coagulation.<br />

Altered ion homeostasis 6 1 2 24 - 1 Deregulation <strong>of</strong> genes encoding proteins involved in ion storage or transport<br />

Energy mobilisation 1 2 1 17 4 3 Upregulation <strong>of</strong> genes primarily involved in glycolysis, fatty acid oxidation or the pentose<br />

phosphate pathway.<br />

Increased transcription and<br />

RNA processing<br />

6 - 2 15 1 6 Upregulation <strong>of</strong> genes encoding transcription factors, coactivators, or enzymes involved in<br />

the metabolism <strong>of</strong> nucleotides and nucleosides and RNA splicing<br />

Increased protein synthesis 1 - 2 4 1 1 Upregulation <strong>of</strong> genes involved in translation or protein folding in the endoplasmatic<br />

reticulum<br />

Increased protein<br />

trafficking<br />

- - 2 28 - - Upregulation <strong>of</strong> genes involved in protein sorting, vesicular transport or endo-/exocytosis<br />

Dedifferentiation 10 2 7 9 9 4 Downregulation <strong>of</strong> genes mediating the physiological homeostasis <strong>of</strong> the cell and/or organ.<br />

Neuronal differentiation 2 2 - 14 1 4 Upregulation <strong>of</strong> genes involved in neuronal signal transmission or neuronal differentiation<br />

Deregulation <strong>of</strong> the<br />

circadian rhythm<br />

Unknown toxicological<br />

category<br />

4 1 - - - - Deregulation <strong>of</strong> genes involved in the regulation <strong>of</strong> the circadian rythm<br />

9 1 2 20 7 12 Genes with known biochemical function, but whose deregulation could not be given a<br />

specific meaning<br />

unknown ESTs 6 1 10 60 16 35 Genes which are not annotated<br />

A B C D E<br />

: ARE: Arylhydrocarbon responsive elements; : NFkB: Nuclear transcription factor B; : Nrf2: NF-E2-related factor-2; : HIF: Hypoxia inducible factor; : (IGF)- PI3K-<br />

AKT: Insulin growth factor- Phosphoinositol-triphosphate kinase-AKT; F : VEGF: Vascular endothelial growth factor; G : TGF-ß: Transforming growth factor ß; H : ECM:<br />

Extracellular matrix; I : TNF: Tumor necrosis factor<br />

64


Chapter 3: Manuscript I<br />

Supplementary table 3.2: Incidence and mean numbers <strong>of</strong> preneoplastic and neoplastic lesions. Mean + SD <strong>of</strong> atypical tubule (AT), aypical hyperplasia (AH)<br />

and neoplastic (adenoma, carcinoma) lesions <strong>of</strong> AA or OTA treated Eker and wild type rats and their respective controls (n=3). Incidences <strong>of</strong><br />

respective lesions in treated or control animals are given in parentheses. Significant differences (unpaired t-test) between treated and timematched<br />

control groups are indicated (*p


Chapter 3: Manuscript I<br />

Supplementary table 3.3: Non-neoplastic pathology <strong>of</strong> AA or OTA treated Eker rats. Mean + SD <strong>of</strong> non-neoplastic changes. ranked according to the following<br />

severity classes: none (0). mild (1). moderate (2). strong (3). and severe (4). including intermediate classes (e.g. 0.5. 1.5) <strong>of</strong> AA or OTA treated<br />

Eker rats and their respective controls (n=3). Incidences <strong>of</strong> respective lesions in treated or control animals are given in parentheses.<br />

Necrosis<br />

Apoptosis<br />

Fibrosis<br />

Basement<br />

membrane<br />

(thickening)<br />

Mesangia<br />

(thickening)<br />

Bowmans Space<br />

with erythrocytes)<br />

Calcium cast<br />

Glomerulo-<br />

Sclerosis<br />

Glomerulo-<br />

Nephritis<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

Control (AA) AA Control (OTA) OTA<br />

d1 d3 d7 d14 d1 d3 d7 d14 d1 d3 d7 d14 d1 d3 d7 d14<br />

0,17 ±<br />

0,24<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.33 ±<br />

0.47<br />

(1/3)<br />

0,17 ±<br />

0,24<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.33 ±<br />

0.47<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.33 ±<br />

0.47<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.3 ±<br />

0.34<br />

(2/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0,17 ±<br />

0,24<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.5 ±<br />

0.71<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.5 ±<br />

0.41<br />

(2/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

Glomeruli<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0,17 ±<br />

0,24<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.33 ±<br />

0.24<br />

(2/3)<br />

0.67 ±<br />

0.47<br />

(2/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0±<br />

0.0<br />

(0/3)<br />

0.33 ±<br />

0.24<br />

(2/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0,17 ±<br />

0,24<br />

(1/3)<br />

0.67 ±<br />

0.24<br />

(3/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.33 ±<br />

0.47<br />

(2/3)<br />

0.83 ±<br />

0.47<br />

(3/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.83 ±<br />

0.24<br />

(3/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0,17 ±<br />

0,24<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.5 ±<br />

0.0<br />

(3/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.5 ±<br />

0.0<br />

(3/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0,17 ±<br />

0,24<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.67 ±<br />

0.47<br />

(2/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

66


Pigment Deposits<br />

Necrosis<br />

Apoptosis<br />

Karyomegally<br />

Vacuolization<br />

Hyaline droplets<br />

Cell shedding<br />

Proteinaceous<br />

casts<br />

Tubular dilatation<br />

Calcium casts<br />

Tubular<br />

regeneration<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

Control (AA) AA Control (OTA) OTA<br />

Chapter 3: Manuscript I<br />

d1 d3 d7 d14 d1 d3 d7 d14 d1 d3 d7 d14 d1 d3 d7 d14<br />

0.33 ±<br />

0.47<br />

(2/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.50 ±<br />

0.41<br />

(2/3)<br />

0.33 ±<br />

0.47<br />

(2/3)<br />

0.67 ±<br />

0.24<br />

(3/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.33 ±<br />

0.24<br />

(2/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.50 ±<br />

0.41<br />

(2/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.33 ±<br />

0.47<br />

(2/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

1.0 ±<br />

0.41<br />

(3/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.50 ±<br />

0.41<br />

(2/3)<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.33 ±<br />

0.47<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.5 ±<br />

0.41<br />

(2/3)<br />

0.33 ±<br />

0.47<br />

(1/3)<br />

0.50 ±<br />

0.41<br />

(2/3)<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.50 ±<br />

0.0<br />

(3/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.33 ±<br />

0.24<br />

(2/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.50 ±<br />

0.41<br />

(2/3)<br />

Cortex<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.33 ±<br />

0.24<br />

(2/3)<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.83 ±<br />

0.24<br />

(3/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.67 ±<br />

0.62<br />

(2/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

1.0 ±<br />

0,41<br />

(3/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.33 ±<br />

0.24<br />

(2/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.33 ±<br />

0.47<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.83 ±<br />

0.47<br />

(3/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.50 ±<br />

0.0<br />

(3/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.83 ±<br />

0.62<br />

(2/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.50 ±<br />

0.0<br />

(3/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

1.83 ±<br />

0.62<br />

(3/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.67 ±<br />

0.47<br />

(2/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.33 ±<br />

0.24<br />

(2/3)<br />

1.5 ±<br />

0.41<br />

(3/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.67 ±<br />

0.47<br />

(3/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.83 ±<br />

0.24<br />

(3/3)<br />

1.0 ±<br />

0.41<br />

(3/3)<br />

0.33 ±<br />

0.47<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

1.33 ±<br />

0.62<br />

(3/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

1.17 ±<br />

0.24<br />

(3/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

1.50 ±<br />

0.41<br />

(3/3)<br />

67


CPN<br />

Inflammation<br />

Hypertrophy<br />

Pigment Deposits<br />

Necrosis<br />

Apoptosis<br />

Karyomegally<br />

Vacuolization<br />

Hyaline droplets<br />

Cell shedding<br />

Proteinaceous<br />

casts<br />

Tubular dilatation<br />

0.33 ±<br />

0.47<br />

(1/3)<br />

0.33 ±<br />

0.47<br />

(1/3)<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

0.33 ±<br />

0.47<br />

(1/3)<br />

0.33 ±<br />

0.47<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

0.33 ±<br />

0.47<br />

(1/3)<br />

0.50 ±<br />

0.41<br />

(2/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.83 ±<br />

0.24<br />

(3/3)<br />

0.33 ±<br />

0.47<br />

(1/3)<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

1.5 ±<br />

0.0<br />

(3/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

Medulla/ Papilla<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

0.50 ±<br />

0.0<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(3/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.33 ±<br />

0.47<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

Chapter 3: Manuscript I<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.50 ±<br />

0.0<br />

(3/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

0.5 ±<br />

0.41<br />

(2/3)<br />

0.67 ±<br />

0.24<br />

(3/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

0.50 ±<br />

0.0<br />

(3/3)<br />

1.0 ±<br />

0.0<br />

(3/3)<br />

0.33 ±<br />

0.47<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

68


Calcium casts<br />

Tubular<br />

regeneration<br />

CPN<br />

Inflammation<br />

Hypertrophy<br />

Chapter 3: Manuscript I<br />

(0/3) (0/3) (0/3) (0/3) (0/3) (0/3) (0/3) (0/3) (0/3) (0/3) (0/3) (0/3) (0/3) (0/3) (0/3) (0/3)<br />

0.33 ±<br />

0.24<br />

(2/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.33 ±<br />

0.24<br />

(2/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.50 ±<br />

0.41<br />

(2/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.33 ±<br />

0.47<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.33 ±<br />

0.34<br />

(2/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.33 ±<br />

0.47<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

1.33 ±<br />

0.94<br />

(2/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.17 ±<br />

0.24<br />

(1/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

0.0 ±<br />

0.0<br />

(0/3)<br />

69


Chapter 3: Manuscript I<br />

Supplementary table 3.4: Non-neoplastic pathology <strong>of</strong> AA or OTA treated wild type rats. Mean + SD <strong>of</strong> non-neoplastic changes, ranked according to the<br />

following severity classes: none (0), mild (1), moderate (2), strong (3), and severe (4), including intermediate classes (e.g. 0.5, 1.5) <strong>of</strong> AA or OTA<br />

treated wild type rats and their respective control (n=3). Incidences <strong>of</strong> respective lesions in treated or control animals are given in parentheses.<br />

Pigment Deposits<br />

Necrosis<br />

Apoptosis<br />

Fibrosis<br />

Basement membrane<br />

(thickening)<br />

Mesangia<br />

Bowmans Space with<br />

erythrocytes)<br />

Calcium cast<br />

Glomerulo- Sclerosis<br />

Glomerulo- Nephritis<br />

Control (AA, OTA) AA OTA<br />

d1 d3 d7 d14 d1 d3 d7 d14 d1 d3 d7 d14<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.17 ±<br />

0.24 (1/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.50 ±<br />

0.71 (1/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.33 ±<br />

0.47 (1/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.33 ±<br />

0.24 (2/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.83 ±<br />

0.62 (2/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.50 ±<br />

0.71 (1/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.5 ± 0.71<br />

(1/3)<br />

0.67 ±<br />

0.62 (2/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

Glomeruli<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.33 ±<br />

0.24 (2/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.50 ±<br />

0.71 (1/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.33 ±<br />

0.24 (2/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

1.0 ± 0.71<br />

(2/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.17 ±<br />

0.24 (1/3)<br />

0.17 ±<br />

0.24 (1/3)<br />

1.0 ± 0.41<br />

(3/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.33 ±<br />

0.24 (1/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.33 ±<br />

0.47 (1/3)<br />

0.17 ±<br />

0.24 (1/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.33 ±<br />

0.24 (2/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.17 ±<br />

0.24 (1/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.17 ±<br />

0.24 (1/3)<br />

0.17 ±<br />

0.24 (1/3)<br />

0.33 ±<br />

0.24 (2/3)<br />

0.67 ±<br />

0.24 (3/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

70


Pigment Deposits<br />

Necrosis<br />

Apoptosis<br />

Karyomegally<br />

Hyaline droplets<br />

Cell shedding<br />

Proteinaceous<br />

casts<br />

Tubular dilatation<br />

Calcium casts<br />

Tubular<br />

regeneration<br />

CPN<br />

Inflammation<br />

Hypertrophy<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.17 ±<br />

0.24 (1/3)<br />

0.17 ±<br />

0.24 (1/3)<br />

0.33 ±<br />

0.47 (1/3)<br />

0.17 ±<br />

0.24 (1/3)<br />

0.33 ±<br />

0.24 (2/3)<br />

0.17 ±<br />

0.24 (1/3)<br />

0.50 ±<br />

0.41 (2/3)<br />

0.33 ±<br />

0.47 (1/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.17 ±<br />

0.24 (1/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.17 ±<br />

0.24 (1/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.33 ±<br />

0.47 (1/3)<br />

0.50 ±<br />

0.41 (2/3)<br />

0.50 ±<br />

0.41 (2/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.17 ±<br />

0.24 (1/3)<br />

1.0 ± 0.0<br />

(3/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.17 ±<br />

0.24 (1/3)<br />

0.17 ±<br />

0.24 (1/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.33 ±<br />

0.47 (1/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.17 ±<br />

0.24 (1/3)<br />

0.83 ±<br />

0.62 (2/3)<br />

0.17 ±<br />

0.24 (1/3)<br />

0.50 ±<br />

0.41 (2/3)<br />

0.50 ±<br />

0.41 (2/3)<br />

0.67 ±<br />

0.24 (3/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.17 ±<br />

0.24 (1/3)<br />

0.33 ±<br />

0.24 (2/3)<br />

0.33 ±<br />

0.47 (1/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.17 ±<br />

0.24 (1/3)<br />

0.33 ±<br />

0.23 (2/3)<br />

0.50 ±<br />

0.41 (2/3)<br />

0.33 ±<br />

0.47 (1/3)<br />

Cortex<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.50 ± 0.0<br />

(3/3)<br />

0.17 ±<br />

0.24 (1/3)<br />

0.67 ±<br />

0.24 (3/3)<br />

0.17 ±<br />

0.24 (1/3)<br />

0.50 ±<br />

0.41 (2/3)<br />

0.17 ±<br />

0.24 (1/3)<br />

0.83 ±<br />

0.24 (3/3)<br />

0.33 ±<br />

0.47 (1/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.50 ±<br />

0.41 (2/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.83 ±<br />

0.62 (2/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.17 ±<br />

0.24 (1/3)<br />

0.17 ±<br />

0.24 (1/3)<br />

0.83 ±<br />

0.24 (3/3)<br />

0.67 ±<br />

0.47 (2/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.17 ±<br />

0.24 (1/3)<br />

0.17 ±<br />

0.24 (1/3)<br />

0.50 ±<br />

0.41 (2/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.17 ±<br />

0.24 (1/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

1.33 ±<br />

0.24 (3/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.17 ±<br />

0.24 (1/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.50 ±<br />

0.41 (2/3)<br />

0.33 ±<br />

0.24 (2/3)<br />

1.17 ±<br />

0.62 (3/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.33 ±<br />

0.24 (2/3)<br />

0.17 ±<br />

0.24 (1/3)<br />

0.33 ±<br />

0.24 (2/3)<br />

0.67 ±<br />

0.47 (2/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.17 ±<br />

0.24 (1/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.33 ±<br />

0.24 (2/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.83 ±<br />

0.85 (2/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.50 ±<br />

0.41 (2/3)<br />

0.33 ±<br />

0.47 (1/3)<br />

0.33 ±<br />

0.47 (1/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

Chapter 3: Manuscript I<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.50 ± 0.0<br />

(3/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.67 ±<br />

0.24 (3/3)<br />

0.33 ±<br />

0.24 (2/3)<br />

0.67 ±<br />

0.47 (2/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.50 ±<br />

0.41 (2/3)<br />

0.67 ±<br />

0.24 (3/3)<br />

0.50 ±<br />

0.41 (2/3)<br />

1.0 ± 0.0<br />

(3/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.17 ±<br />

0.24 (1/3)<br />

0.83 ±<br />

0.24 (3/3)<br />

0.50 ± 0.0<br />

(3/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.67 ±<br />

0.47 (2/3)<br />

0.17 ±<br />

0.24 (1/3)<br />

1.0 ± 0.0<br />

(3/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.83 ±<br />

0.85 (2/3)<br />

0.50 ±<br />

0.41 (2/3)<br />

0.83 ±<br />

0.24 (3/3)<br />

0.33 ±<br />

0.47 (1/3)<br />

71


Pigment Deposits<br />

Necrosis<br />

Apoptosis<br />

Karyomegally<br />

Vacuolization<br />

Hyaline droplets<br />

Cell shedding<br />

Proteinaceous<br />

casts<br />

Tubular dilatation<br />

Calcium casts<br />

Tubular<br />

regeneration<br />

CPN<br />

Inflammation<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.33 ± 0.47<br />

(1/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.17 ± 0.24<br />

(1/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.17 ± 0.24<br />

(1/3)<br />

0.17 ± 0.24<br />

(1/3)<br />

Medulla/ Papilla<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.33 ± 0.47<br />

(1/3)<br />

0.33 ± 0.47<br />

(1/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

Chapter 3: Manuscript I<br />

0.0 ± 0.0<br />

(0/3)<br />

0.33 ± 0.47<br />

(1/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

0.0 ± 0.0<br />

(0/3)<br />

72


Chapter 4: Manuscript II<br />

Renal Carcinogenic Response <strong>of</strong> Eker Rats to Low Doses <strong>of</strong><br />

Methylazoxymethanol Acetate<br />

Kerstin Stemmer 1 , Heidrun Ellinger-Ziegelbauer 2 , Katja Strauch 3 , Leonard Collins 3 , James A.<br />

Swenberg 3 , Hans-J. Ahr 2 and Daniel R. Dietrich 1*<br />

1<br />

Human and Environmental Toxicology, University <strong>of</strong> Konstanz, Konstanz, Germany<br />

2<br />

<strong>Molecular</strong> and Special Toxicology, Bayer Healthcare AG, Wuppertal, Germany<br />

3<br />

Environmental Sciences and Engineering, University <strong>of</strong> North Carolina at Chapel Hill, Chapel<br />

Hill, North Carolina, USA<br />

* Corresponding author: Daniel R. Dietrich: daniel.dietrich@uni-konstanz.de<br />

Submitted to Mutation Research<br />

4.1 Abstract<br />

Methylazoxymethanol-actetae (MAMAc), the stabilized aglycone <strong>of</strong> cycasin, is a potent<br />

renal carcinogen in rodents, which acts via direct alkylation <strong>of</strong> the DNA. Current knowledge<br />

on MAMAc carcinogenicity is based on the use <strong>of</strong> very high doses, which may reflect a<br />

carcinogenic mechanism different from the one active at low doses. This study compared<br />

the renal response <strong>of</strong> male and female Tsc2-mutant Eker rats exposed to low doses <strong>of</strong><br />

MAMAc (250µg/kg bw day -1 ) after 1, 3, 7 and 14 days <strong>of</strong> exposure using gene expression<br />

pr<strong>of</strong>iling, histopathology, including the assessment <strong>of</strong> preneoplastic and neoplastic lesions,<br />

cell proliferation analysis and LC-MS/MS for demonstration <strong>of</strong> O6-methylguanine (O6-meG)<br />

DNA adducts. Following 3 and 6 months <strong>of</strong> exposure renal histopathology, including<br />

preneoplastic and neoplastic lesions and cell proliferation was assessed.<br />

Gene expression pr<strong>of</strong>iles revealed only a small number <strong>of</strong> deregulated genes in short-term<br />

MAMAc treated males, which mainly revealed a reduced protein synthesis, including downregulation<br />

<strong>of</strong> tumor suppressor genes. An exposure dependent increase in O6-meG<br />

adducts was indicated following 1-14 days <strong>of</strong> exposure, whereas the number <strong>of</strong> O6-meG<br />

adducts detected were generally higher in female than in male rats. 250µg/kg bw day -1 <strong>of</strong><br />

73


Chapter 4: Manuscript II<br />

MAMAc for 3 and 6 months induced increased cell proliferation and number <strong>of</strong><br />

preneoplastic lesions only in female but not in male rats. However, surprisingly male Eker<br />

rats presented with an increased incidence and total higher number <strong>of</strong> adenomas and<br />

carcinomas compared to female rats after 6 months <strong>of</strong> exposure.<br />

In conclusion, above findings suggests that subchronic and –chronic exposure to low doses<br />

<strong>of</strong> MAMAc results in gender-specific differences in response. Indeed, in female rats the<br />

increase in early tumor stages and absence <strong>of</strong> late stages points to a tumor initiating role <strong>of</strong><br />

MAMAc, while in male rats, the increase in later tumor stages after 6 months exposure is<br />

suggestive <strong>of</strong> a tumor promoting role <strong>of</strong> MAMAc.<br />

4.2 Introduction<br />

Cycasin (methylazoymethanol-ß-D-glucoside) is a natural constituent <strong>of</strong> nuts, root and<br />

leaves <strong>of</strong> the cycad plants Cycas circinalis and Cycas revoluta that have been sources <strong>of</strong><br />

food and medicine for the indigenous people <strong>of</strong> the Western Pacific, e.g. the island <strong>of</strong><br />

Guam (Whiting 1963). Cycasin contamination mainly depend on food procession, for<br />

instance flour from washed cycad nuts from Guam only contain marginal concentrations,<br />

while up to 2.3% cycasin was detected when unwashed nuts were used (Hirono 1981). In<br />

rats (Laqueur 1964; Laqueur and Matsumoto 1966), mice (O'Gara et al. 1964), guinea pigs<br />

(Spatz 1964), rabbits (Hirono 1972) and nonhuman primates (Sieber et al. 1980), cycasin<br />

was found to be a potent carcinogen resulting primarily in epithelial and mesenchymal<br />

tumors <strong>of</strong> the kidneys, tumors <strong>of</strong> the liver and colon.<br />

The carcinogenic action <strong>of</strong> cycasin appears mainly dependent on its deglucosylation by ßglucosidases<br />

<strong>of</strong> the gut micr<strong>of</strong>lora to release its principal metabolite methylazoxymethanol<br />

(MAM) (Spatz et al. 1967; Matsushima et al. 1979; Choi et al. 1996). The release <strong>of</strong> the<br />

“ultimate carcinogen” methyldiazonium ion from MAM (Figure 4.1) is either catalyzed by<br />

different enzymes including alcohol dehydrogenase (Grab and Zedeck 1977; Notman et al.<br />

1982), choline dehydrogenase (Tan et al. 2004) and cytochrome P450-2E1 (Sohn et al.<br />

2001), however, may also occur spontaneously under normal physiological conditions<br />

(Nagasawa et al. 1972; Sohn et al. 2001). The methyldiazonium ion, is also metabolically<br />

formed from other DNA alkylating pro-carcinogens, e.g. dimethylnitrosamine (DMN) (Shank<br />

and Magee 1967; Weisburger 1978) and was demonstrated to be responsible for the<br />

formation <strong>of</strong> pro-mutagenic and pro-carcinogenic N7-methylguanine- and O6-<br />

74


Chapter 4: Manuscript II<br />

methylguanine- DNA- adducts observed in different target organs <strong>of</strong> rats, treated with DNA<br />

alkylating pro-carcinogens (Matsumoto and Higa 1966; Sohn et al. 2001).<br />

C<br />

H 3<br />

N +<br />

O -<br />

I<br />

N<br />

CH2 O<br />

O<br />

OH<br />

CH 2 OH<br />

OH OH<br />

β-glucosidase<br />

Glycoside<br />

C<br />

H 3<br />

N + N<br />

IV<br />

O -<br />

H<br />

C O<br />

enzymatic<br />

C<br />

H 3<br />

N + N<br />

DNA<br />

O -<br />

H<br />

CH2 O<br />

spontaneous<br />

II<br />

C<br />

H 3<br />

nucleophilic<br />

attack<br />

C<br />

H 2<br />

V<br />

N +<br />

III<br />

O<br />

+<br />

H3C +<br />

VI<br />

N<br />

+ OH -<br />

Figure 4.1: Metabolic pathways <strong>of</strong> cycasin and MAM activation, from (Laqueur 1968; Nagasawa et<br />

al. 1972; Sohn et al. 2001). I: cycasin; II: MAM; III: formaldehyde; IV:<br />

methylazoxyformaldehyde; V: methyldiazonium ion, VI: methylcarbonium ion.<br />

However, since earlier cycasin and MAM carcinogenicity studies used high oral doses (4-<br />

500mg kg-1 bw), either as single or as multiple applications over a short exposure period<br />

(Hirono et al. 1968; Laqueur 1968; Fukunishi et al. 1971), it remains unclear whether the<br />

exposure to acute, subchronic or chronic low doses <strong>of</strong> MAM is sufficient to induce initiate<br />

and/or promote carcinogenic lesions or other adverse effects in the rat kidney.<br />

To elucidate this question, a sensitive rat model for the detection <strong>of</strong> renal carcinogens was<br />

employed: Eker rats carrying an inherited heterozygous mutation in the tuberous sclerosis<br />

(TSC2) tumor suppressor gene (Yeung et al. 1994; Hino et al. 1995; Kobayashi et al. 1997)<br />

are predisposed to develop renal cancer early <strong>of</strong> age and were previously characterized to<br />

be highly sensitive towards renal carcinogens (McDorman and Wolf 2002; Hino 2004). For<br />

instance, when treated with a single dose <strong>of</strong> the DNA alkylating DMN (30mg/kg bw ), 8<br />

months later, Eker rats demonstrated a 70-fold higher average number <strong>of</strong> tumors per<br />

animal when compared to the corresponding wild type rats exposed under identical<br />

conditions (Walker et al. 1992 8).<br />

Based on above findings it was expected that Eker rats, continuously exposed to low doses<br />

<strong>of</strong> MAM-acetate (MAMAc, 250µg/kg bw), should present with a detectable increase <strong>of</strong> preneoplastic<br />

and neoplastic lesions already following 6 months <strong>of</strong> exposure. Gene-expression<br />

pr<strong>of</strong>iling was previously demonstrated to be a sensitive and reliable tool that allows<br />

N 2<br />

75


Chapter 4: Manuscript II<br />

detection <strong>of</strong> the activation <strong>of</strong> a specific mode <strong>of</strong> action <strong>of</strong> renal or liver carcinogens (toxicity<br />

and carcinogenicity) already after 1 to 14 days <strong>of</strong> carcinogen exposure in Eker (Stemmer et<br />

al. 2007) or Wistar rats (Ellinger-Ziegelbauer et al. 2004; Ellinger-Ziegelbauer et al. 2005),<br />

respectively. Indeed, an up-regulation <strong>of</strong> genes involved in the DNA damage response was<br />

reported in livers <strong>of</strong> DMN-treated Wistar rats after one single dose <strong>of</strong> 4mg/kg bw (Ellinger-<br />

Ziegelbauer et al. 2004). Thus, gene expression pr<strong>of</strong>iling, subsequent to 1-14 days low<br />

dose MAMAc treatment <strong>of</strong> Eker rats, should allow the detection <strong>of</strong> a possible DNA damage<br />

response as well as other specific responses associated with the toxicity and/or the<br />

carcinogenicity <strong>of</strong> MAMAc. The latter in combination with renal pathology and cell<br />

proliferation analysis would therefore allow the determination <strong>of</strong> the acute toxic effects and<br />

earliest events <strong>of</strong> MAMAc induced renal tumorigenesis, whereas renal histopathology,<br />

including the assessment <strong>of</strong> preneoplastic and neoplastic lesions, and cell proliferation<br />

analysis following 3 and 6 months <strong>of</strong> exposure would allow to determine whether<br />

subchronic and chronic low dose MAMAc treatment <strong>of</strong> Eker rats manifests in the onset <strong>of</strong><br />

preneoplastic and neoplasic renal lesions. Possible sex-specific effects as observed for<br />

other renal carcinogens (Boorman et al. 1992) were addressed via employing male and<br />

female Eker rats.<br />

4.3 Material and Methods<br />

4.3.1 Compound<br />

Highly purified methylazoxymethanol acetate (MAMAc) was purchased from the Midwest<br />

Research Institute, NCI Chemical Resource Repository (64 FR 72090, 64 FR 28205). Prior<br />

to the in vivo experiments, the stability <strong>of</strong> MAMAc in 0.1M sodium bicarbonate (NaHCO3)<br />

was confirmed by HPLC using a method published previously (Fiala et al. 1976). Stabilized<br />

MAMAc is highly sensitive to hydrolysis by various blood and organ esterases, and<br />

therefore is deacetylated to MAM almost immediately following administration (Zedeck and<br />

Brown 1977)<br />

4.3.2 Animals<br />

Six to ten week old, genotyped heterozygous Tsc2 mutant Eker rats (Tsc2+/-, Long Evans)<br />

were purchased from the MD Anderson <strong>Cancer</strong> Center, Smithville, Texas, USA and housed<br />

at the University <strong>of</strong> Konstanz animal research facility under standard conditions. Prior to<br />

76


Chapter 4: Manuscript II<br />

exposure, male and female Eker rats were randomly assigned to dose groups and allowed<br />

to acclimatize to laboratory conditions for 4 weeks.<br />

4.3.3 Short-term experiments<br />

Groups <strong>of</strong> three male and female Eker rats were gavaged daily with MAMAc (250µg/ kg<br />

BW) dissolved in sterile 0.1M NaHCO3 for 1, 3, 7, and 14 days, respectively (Table 4.1).<br />

Time-matched vehicle controls (n=3) were gavaged with corresponding volumes <strong>of</strong> 0,1M<br />

NaHCO3.<br />

4.3.4 Subchronic and chronic experiments<br />

Groups <strong>of</strong> 10 Male and female Eker rats were gavaged with MAMAc (250µg/ kg BW),<br />

dissolved in 0.1M NaHCO3 at five days a week for 3 and 6 months. Time-matched vehicle<br />

controls were gavaged with 0.1M NaHCO3. For post-mortem immunohistochemical<br />

analysis <strong>of</strong> cell proliferation, 5 <strong>of</strong> 10 rats per dose group and sex were s.c. implanted with<br />

osmotic ALZET-pumps (Model 2ML1, Charles River Laboratories, Germany) containing 5bromo-2-deoxyuridine<br />

(BrdU, 20mg/ml sterile saline, Sigma Aldrich, Germany) 5 days prior<br />

to sacrifice.<br />

4.3.5 Sample collection<br />

At the end <strong>of</strong> each treatment period, anesthetized rats were sacrificed by exsanguination<br />

subsequent to retrograde perfusion with PBS and kidneys were collected, longitudinally<br />

sectioned into 5mm slices and stored in RNAlater (Qiagen, Germany) or in PBS buffered<br />

histology fixative buffer, containing 2% paraformaldehyde and 1% glutaraldehyde for<br />

subsequent paraffin embedding and sectioning.<br />

4.3.6 Histopathology<br />

Haematoxilin and Eosin (H&E) stained renal paraffin section were randomized and<br />

histopathological analysis was carried out by light microscopy at 40-400-fold magnification.<br />

Non-neoplastic changes were classified as none (0), mild (1), moderate (2), strong (3), and<br />

severe (4), including intermediate classes (e.g. 0.5, 1.5 etc.), while preneoplastic and<br />

77


Chapter 4: Manuscript II<br />

neoplastic lesions were classified as reported previously (Dietrich and Swenberg 1991).<br />

Absolute numbers and incidences <strong>of</strong> preneoplastic and neoplastic lesions were determined.<br />

4.3.7 Cell proliferation analysis<br />

Immunostaining: Subsequent to short term experiments (1-14 days), cell proliferation was<br />

evaluated by immunohistochemical staining for proliferating cell nuclear antigen (PCNA)<br />

using monoclonal primary anti-PCNA antibody (PC-10; DAKO, Germany) in paraffinembedded<br />

kidney sections, as described previously (Stemmer et al. 2007).<br />

Five days prior to the sub-chronic (3 months) and chronic (6 months) experiments 5 <strong>of</strong> 10<br />

rats per dose group and sex were s.c. implanted with osmotic ALZET-pumps (Model 2ML1,<br />

Charles River Laboratories, Germany) containing 5-bromo-2-deoxyuridine (BrdU, 20mg/ml<br />

sterile saline, Sigma Aldrich, Germany) to allow post-mortem immunohistochemical<br />

analysis <strong>of</strong> cell proliferation. BrdU-immunostaining was performed as described previously<br />

(Stemmer et al. 2007), using a monoclonal mouse anti-BrdU primary antibody (MU247-UC,<br />

Biogenex, USA) diluted 1:100 in Power BlockTM (BioGenex, USA) in an overnight<br />

application at 4°C.<br />

Quantification <strong>of</strong> S-phase nuclei: Subsequent to short-term, sub-chronic and chronic<br />

experiments, PCNA- or BrdU- positive stained S-phase nuclei were quantified on<br />

randomized sections. 20 microscopic fields (10x ocular, 40x objective) were randomly<br />

chosen within the area <strong>of</strong> the renal outer cortex and inner cortex/outer medulla. Proximal<br />

and distal tubules and collecting ducts were counted separately per field. A minimum <strong>of</strong><br />

1.000 and 500 nuclei were counted in proximal and distal tubules and collecting ducts,<br />

respectively, distinguishing between negative and positive BrdU or PCNA-stained nuclei.<br />

Nuclear labeling indices (LI%) were calculated as positive nuclei/total number <strong>of</strong> nuclei<br />

counted.<br />

4.3.8 RNA isolation microarray hybridization and gene expression<br />

pr<strong>of</strong>iling<br />

RNA isolation from RNAlater fixed kidneys and Affymetrix Rat Genome RAE230A array<br />

hybridization was performed as described previously (Stemmer et al. 2007). Microarray<br />

quality control was performed as published by Elling-Ziegelbauer (Ellinger-Ziegelbauer et<br />

al. 2004). Statistical analysis was performed using Expressionist Analyst s<strong>of</strong>tware<br />

(Genedata AG, Switzerland). To select significantly deregulated genes due to treatment or<br />

78


Chapter 4: Manuscript II<br />

time (single effects) and to treatment and time (interaction effect), 2-way ANOVA with a pvalue<br />

cut-<strong>of</strong>f <strong>of</strong> 0.005, combined with a 1.7-fold deregulation threshold for at least one timepoint<br />

was applied. Significantly deregulated genes were subsequently divided into gene<br />

groups with distinct expression pr<strong>of</strong>iles over the time-course using self-organizing map<br />

(SOM) analysis. SOM analysis also allowed the identification <strong>of</strong> genes showing inconsistent<br />

expression between the controls at different time points. Such genes were manually<br />

discarded from the subsequent functional analysis. Using this adjusted dataset, gene<br />

expression ratios <strong>of</strong> individual genes were calculated by dividing the respective expression<br />

values <strong>of</strong> single treated replicate samples by the mean expression value <strong>of</strong> all<br />

corresponding time-matched control samples. Heat-maps were used to graphically display<br />

the relative expression data, after one-dimensional clustering <strong>of</strong> the genes.<br />

Significantly deregulated genes were characterized according to the physiological role <strong>of</strong> its<br />

encoded protein, using information from databases, e.g. NetAffx (update: August 2006),<br />

Swissprot, Proteome and Pubmed.<br />

4.3.9 Quantification <strong>of</strong> O6MG<br />

DNA was isolated and extracted from pooled kidney samples <strong>of</strong> three replicate animals<br />

using an ABI 340A nucleic acid extractor with two phenol/chlor<strong>of</strong>orm and one chlor<strong>of</strong>orm<br />

extraction, followed by precipitation with sodium acetate and isopropanol. DNA adduct<br />

formation in kidneys <strong>of</strong> MAMAc-treated rats was measured by liquid chromatography<br />

coupled with mass spectrometry. The actual analyses were conducted under a pay-for<br />

service contract in the analytical core facility <strong>of</strong> the Environmental Sciences and<br />

Engineering, University <strong>of</strong> North Carolina at Chapel Hill, USA, using undisclosed<br />

methodology.<br />

4.3.10 Statistical analysis<br />

Statistical analysis <strong>of</strong> histopathological and cell proliferation data were carried out using<br />

GraphPad Prism 4® S<strong>of</strong>tware. Significant differences in nuclear labeling indices (LI %) or<br />

total number <strong>of</strong> lesions in treated and control rats were analyzed by Bonferroni’s test for<br />

multiple comparisons. Lesion incidences were tested for significance using the 2-sided<br />

Fisher’s exact test. Ranked non-neoplastic pathology data were analyzed using the<br />

nonparametric Mann-Whitney test.<br />

79


4.4 Results<br />

4.4.1 Gene expression pr<strong>of</strong>iling<br />

Chapter 4: Manuscript II<br />

Oral treatment <strong>of</strong> male Eker rats with MAMAc for 1, 3, 7 and 14 days resulted in only a<br />

small number <strong>of</strong> significantly deregulated genes in the renal cortex. From overall 59<br />

significantly deregulated annotated genes 15 were down-regulated. Many <strong>of</strong> the up- and<br />

down-regulated genes demonstrated a transient deregulation, with highest gene expression<br />

values after one day <strong>of</strong> exposure (Figure 4.2).<br />

Figure 4.2: Heat map <strong>of</strong> compound-specific unions <strong>of</strong> genes found significantly deregulated (red:<br />

up-regulated; green: down-regulated) from the corresponding control in MAMAc<br />

induced renal gene expression pr<strong>of</strong>iles <strong>of</strong> Eker after 1, 3, 7 or 14 days <strong>of</strong> treatment<br />

(n=3 per time-point). Color scale (left side): gene expression ratio.<br />

Interpretation <strong>of</strong> significantly deregulated genes according to their physiological role and<br />

their direction <strong>of</strong> deregulation allowed categorization <strong>of</strong> genes into different functional<br />

classes. MAMAc treatment resulted in a deregulation <strong>of</strong> two prominent gene clusters<br />

pointing to a reduced transcription and protein synthesis and an up-regulation <strong>of</strong> the TGFß<br />

pathway. An overview <strong>of</strong> these categories is given in table 4.1, while specific responses are<br />

detailed below. Other clusters included genes involved in ion homeostasis, intracellular<br />

transport and regulation <strong>of</strong> the cytoskeleton. Importantly, MAMAc treatment did not result in<br />

genes encoding proteins involved in DNA damage response (Table 4.1 and below).<br />

80


Chapter 4: Manuscript II<br />

Table 4.1: Categories <strong>of</strong> genes differentially deregulated by MAMAc in Eker- and wild type rats. For the major genes cluster are listed together with their<br />

Genebank accession number and their main biochemical function. For Eker- and wild type rats, the fold deregulation ratios <strong>of</strong> genes that were<br />

significantly deregulated at least at on time point according to N-way ANOVA are indicated bold in the last four columns.<br />

Cluster Accession<br />

number<br />

Reduced<br />

transcription<br />

Gene title Biochemical category Fold deregulation (mean <strong>of</strong> three replicate animals<br />

per time-point vs. mean controls)<br />

AI235236 H2AFO H2A histone family member O Regulation <strong>of</strong> gene expression /<br />

chromatin assembly<br />

BF398015 CBX5 chromobox homolog 5 (Drosophila HP1a) Regulation <strong>of</strong> gene expression/<br />

chromatin assembly<br />

BI300772 RING1 Ring finger protein 1 Regulation <strong>of</strong> gene expression/<br />

chromatin assembly<br />

M25804 NR1D1 Nuclear receptor subfamily 1D1 (Rev-ErbAalpha)<br />

BF284190 NR1D2 Nuclear receptor subfamily 1D2 (Rev-ErbAbeta)<br />

BF420722 NFIX Nuclear factor I-X (CCAAT-binding<br />

transcription factor)<br />

Regulation <strong>of</strong> gene expression<br />

(Transcription factor)<br />

Regulation <strong>of</strong> gene expression<br />

(Transcription factor)<br />

Regulation <strong>of</strong> gene expression<br />

(Transcription factor)<br />

AI008883 FKHL18 Forkhead-like 18 (Drosophila) [predicted] Regulation <strong>of</strong> gene expression<br />

(Transcription factor)<br />

NM_012760 PLAGL1 Pleiomorphic adenoma gene-like 1 Regulation <strong>of</strong> gene expression<br />

(Transcription factor)<br />

1387874_at DBP D-site albumin promoter binding protein 1 Regulation <strong>of</strong> gene expression<br />

(Transcription factor)<br />

NM_133303 BHLHB3 Basic helix-loop-helix protein B3 Regulation <strong>of</strong> gene expression<br />

(Transcription factor)<br />

BG371810 BAT1A ATP-depende nt RNA helicase P4<br />

(Spliceosome RNA helicase Bat1)<br />

NM_053849 PDIA4 Protein disulfide isomerase A4 (CABP2<br />

Calcium-binding protein 2)<br />

Ek d1 Ek d3 Ek d7 Ek d14<br />

0.49 + 0.02 0.64 + 0.06 0.94 + 0.02 0.79 + 0.14<br />

0.79 + 0.20 0.72 + 0.10 0.65 + 0.04 0. 56 + 0.21<br />

0.52 + 0.17 0.83 + 0.13 0.94 + 0.07 0.69 + 0.12<br />

0.57 + 0.10 0.74 + 0.13 0.51 + 0.04 0.33 + 0.08<br />

0.66 + 0.11 0.74 + 0.02 0.45 + 0.02 0.32 + 0.07<br />

0.22 + 0.00 0.28 + 0.01 0.80 + 0.21 0.85 + 0.10<br />

0.94 + 0.07 0.50 + 0.10 1.32 + 0.25 0.73 + 0.18<br />

0.58 + 0.07 0.80 + 0.09 0.86 + 0.05 0.90 + 0.15<br />

0.53 + 0.16 0.94 + 0.09 0.81 + 0.12 1.06 + 0.03<br />

0.56 + 0.15 1.06 + 0.18 0.97 + 0.10 0.99 + 0.07<br />

RNA metabolism (Splicing) 0.88 + 0.03 0.74 + 0.03 0.48 + 0.00 1.10 + 0.07<br />

Protein metabolism (Folding) 0.67 + 0.07 0.47 + 0.05 0.61 + 0.07 0.68 + 0.05<br />

81


TGFß pathway<br />

activation<br />

DNA-damage<br />

response<br />

Accession<br />

number<br />

Chapter 4: Manuscript II<br />

Gene title Biochemical category Fold deregulation (mean <strong>of</strong> three replicate animals<br />

per time-point vs. mean controls)<br />

NM_012775 TGFBR1 TGF-beta receptor type I (ALK5) Signaling cascades (TGF-ß<br />

family pathway)<br />

L09653 TGFBR2 TGF-beta receptor type II Signaling cascades (TGF-ß<br />

family pathway)<br />

NM_013095 SMAD3 (MADH3 Mothers against decapentaplegic<br />

homolog 3)<br />

NM_020085 PTPRK Protein tyrosine phosphatase, (receptortype,<br />

kappa), extracellular region<br />

Signaling cascades (TGF-ß<br />

family pathway)<br />

Signaling cascades (TGF-ß<br />

family pathway)<br />

Ek d1 Ek d3 Ek d7 Ek d14<br />

1.97 + 0.13 1.66 + 0.19 1.32 + 0.18 1.15 + 0.19<br />

5.07 + 0.79 1.33 + 0.27 3.75 + 1.45 2.08 + 0.73<br />

2.06 + 0.21 1.44 + 0.15 1.06 + 0.11 1.17 + 0.19<br />

2.79 + 0.79 2.78 + 0.30 1.73 + 0.73 1.11 + 0.30<br />

NM_012861 MGMT O-6-methylguanine-DNA methyltransferase DNA damage repair 0.86 + 0.79 0.97 + 0.03 1.03 + 0.17 0.91 + 0.06<br />

NM_012601 MPG N-methylpurine-DNA glycosylase DNA damage repair (BER) 1.15 + 0.11 1.03 + 007 1.14 + 0.04 1.18 + 0.15<br />

AF311054<br />

NM_017141<br />

NM_021662<br />

APEX1 apurinic/apyrimidinic endonuclease 1 DNA damage repair (BER) 1.04 + 0.20 0.79 + 0.20 0.79 + 0.34 0.99 + 0.11<br />

POLB Polymerase (DNA directed). beta DNA damage repair (BER) 1.02 + 0.06 0.96 + 0.09 0.84 + 0.07 0.83 + 0.08<br />

POLD polymerase (DNA directed). delta DNA damage repair (BER) 0.92 + 0.12 1.06 + 0.34 1.11 + 0.22 1.11 + 0.35<br />

82


Chapter 4: Manuscript II<br />

DNA damage response: Due to the known genotoxic action <strong>of</strong> MAMAc, special focus was<br />

set on genes coding for proteins that could be indicative for a MAMAc induced DNA<br />

damage. However, genes involved in the expected O6meG adduct repair, including O6methylguanine-DNA<br />

methyltransferase (MGMT) and enzymes <strong>of</strong> the base excision repair<br />

pathway involving N-methylpurine-DNA glycosylase (MPG), apurinic endonuclease<br />

(APEX), DNA polymerase ß and δ (Sedgwick et al. 2007) were not up-regulated in MAMAc<br />

treated male rats (Table 4.1). Deregulating <strong>of</strong> other genes involved in DNA damage repair,<br />

cell cycle arrest or apoptosis could also not be detected (data not shown).<br />

Reduced mRNA synthesis: The most prominent deregulated gene cluster detectable after<br />

MAMAc treatment includes 11 <strong>of</strong> the overall 15 down-regulated genes point to a decreased<br />

overall mRNA synthesis, including down-regulated genes involved in chromatin remodeling<br />

(H2A, CBX5, RING1), basic transcription factors (e.g. PLAGL1, NR1D1, NR1D2, NFIX,<br />

FKHL18, DBP, BHLHB3), RNA splicing and translation (BAT1A).<br />

Signaling cascades: Up-regulated genes and gene-clusters following MAMAc treatment<br />

included 4 genes involved in TGFß signaling (TGFBR1, TGFBR2, SMAD3 and PTPRK),<br />

see table 4.1.<br />

4.4.2 Non-neoplastic renal pathology<br />

Up to 6 months treatment with MAMAc did not result in overt renal non-neoplastic<br />

pathology in male or female Eker rats when compared to the respective controls<br />

(Supplementary tables 4.1 and 4.2). Both MAMAc treated and control rats <strong>of</strong> either sex<br />

presented with minimal renal pathology i.e. cell shedding, tubular dilatation, protein casts,<br />

tubular regeneration and chronic progressive nephropathy (Table 4.2). Notably, epithelial<br />

vacuolar degeneration in the corticomedullary was observed exclusively in MAMAc treated<br />

and control female rats, while this was absent in the corresponding male rats.<br />

83


Chapter 4: Manuscript II<br />

Table 4.2: Non-neoplastic renal pathology <strong>of</strong> male and female Eker rats treated with MAMAc for<br />

three and six months respectively. Histopathological changes were ranked from none<br />

(0) to severe (4) including intermediate classes. Values are presented as median ±<br />

median absolute deviation.<br />

3 months 6 months<br />

males females males females<br />

Control MAMAc Control MAMAc Control MAMAc Control MAMAc<br />

Necrosis 0.0 + 0.0 0.0 + 0.0 0.0 + 0.0 0.0 + 0.0 0.0 + 0.0 0.0 + 0.0 0.0 + 0.3 0.5 + 0.3<br />

Apoptosis 0.0 + 0.0 0.0 + 0.0 0.0 + 0.0 0.0 + 0.1 0.0 + 0.0 0.0 + 0.0 0.0 + 0.0 0.0 + 0.0<br />

Karyomegaly 0.0 + 0.0 0.0 + 0.0 0.0 + 0.1 0.0 + 0.1 0.0 + 0.0 0.0 + 0.0 0.0 + 0.0 0.0 + 0.1<br />

Vacuolization 0.0 + 0.0 0.0 + 0.1 0.0 + 0.2 0.0 + 0.0 0.0 + 0.0 0.0 + 0.2 2.0 + 0.4 1.5 + 0.6<br />

Cell shedding 1.0 + 0.6 0.8 + 0.3 1.0 + 0.3 0.5 + 0.3 1.0 + 0.6 0.5 + 0.3 1.0 + 0.4 1.0 + 0.3<br />

Protein casts 0.3 + 0.6 0.3 + 0.3 0.5 + 0.5 0.5 + 0.4 1.0 + 0.5 1.3 + 0.6 0.0 + 0.7 1.0 + 0.7<br />

Tubular<br />

dilatation<br />

0.5 + 0.5 0.5 + 0.2 1.0 + 0.6 0.8 + 0.4 0.5 + 1.0 1.0 + 0.3 0.5 + 0.5 0.5 + 0.3<br />

Ca 2+ Casts 0.0 + 0.4 0.0 + 0.1 0.0 + 0.2 0.0 + 0.4 0.0 + 0.0 0.0 + 0.2 0.0 + 0.0 0.0 + 0.2<br />

Regeneration 0.5 + 0.2 0.5 + 0.3 0.3 + 0.4 0.5 + 0.2 0.0 + 0.5 1.0 + 0.3 0.5 + 0.5 0.5 + 0.4<br />

CPN 0.5 + 0.2 0.5 + 0.2 0.5 + 0.2 0.5 + 0.4 0.8 + 0.5 1.0 + 0.3 1.0 + 0.3 0.8 + 0.4<br />

Inflammation 0.3 + 0.3 0.0 + 0.1 0.5 + 0.2 0.0 + 0.1 0.5 + 0.4 0.3 + 0.3 0.5 + 0.3 0.0 + 0.3<br />

Pigment<br />

deposit<br />

0.1 + 0.2 0.0 + 0.1 0.1 + 0.2 0.0 + 0.1 0.0 + 0.1 0.0 + 0.2 0.0 + 0.3 0.0 + 0.1<br />

4.4.3 (Pre)-neoplastic renal pathology<br />

Histopathological analysis demonstrated the occurrence <strong>of</strong> preneoplastic and neoplastic<br />

lesions <strong>of</strong> different progressional stages, i.e. basophilic atypical tubules (bATs), basophilic<br />

hyperplasia (bAHs), adenomas and carcinomas <strong>of</strong> a basophilic phenotype in all groups<br />

examined. Representative images <strong>of</strong> preneoplastic and neoplastic lesions are shown in<br />

Figure 4.3, A-C.<br />

One to 14 days MAMAc treatment did not result in a significant increase <strong>of</strong> preneoplastic<br />

and neoplastic lesions in either sex. Yet a clear, but non-significant increase in total bAT<br />

numbers were found in 14 days (Figure 4.4, A and B).<br />

84


Chapter 4: Manuscript II<br />

Figure 4.3: Representative preneoplastic lesions as observed in H&E stained renal sections (A)<br />

bAT; (B) bAH, (C) carcinoma, (D) BrdU staining.<br />

Although a 100% incidence <strong>of</strong> bATs was found in all treatment and control groups after<br />

three and six months <strong>of</strong> exposure, no significant increase <strong>of</strong> absolute numbers could be<br />

detected in MAMAc treated males, when compared to the respective controls (Figure 4.4, C<br />

and D). Only female Eker rats presented with a significant 1.7-fold increase <strong>of</strong> bATs after<br />

three months <strong>of</strong> MAMAc treatment (Figure 4.4, C). However, the latter effect appeared to<br />

be transient as it was not seen in the 6 month treated females (Figure 4.4, D). A higher yet<br />

non-significant total number and incidence <strong>of</strong> microscopically detected adenomas and<br />

carcinomas was present in 6 months MAMAc treated male than in female rats, when<br />

corrected for the numbers and incidences observed in the respective controls.<br />

85


Chapter 4: Manuscript II<br />

Figure 4.4: Mean number <strong>of</strong> preneoplastic lesions in male and female Eker rats after 1, 3, 7 and 14 days (A-B) or 3 and 6 month (C-D) <strong>of</strong> MAMAc exposure<br />

compared to the respective control groups.Light grey bars: bAT; white bars: bAHs, black.<br />

86


4.4.4 Cell proliferation<br />

Chapter 4: Manuscript II<br />

Site-specific cell proliferation analysis was assessed via PCNA and BrdU immunostaining<br />

(Figure 4.3, D) after acute and chronic MAMAc treatment respectively. In general nuclei<br />

with positive PCNA staining were rare (below 1% <strong>of</strong> all nuclei counted) and mainly localized<br />

in proximal tubules <strong>of</strong> the inner and outer cortex. As demonstrated in figure 4.5, A, 14 days<br />

treatment with MAMAc resulted in no significant change in cell proliferation rate in the<br />

proximal tubules in the outer or inner cortex <strong>of</strong> male Eker rats. In contrast 14 days<br />

treatment <strong>of</strong> females with MAMAc resulted in a significant 1.5 fold increase <strong>of</strong> cell<br />

proliferation in the inner cortex compared to the time-matched controls (Figure 4.5, B).<br />

Figure 4.5: (A-B) PCNA S-Phase mean labeling indices (LI %) in the outer and inner renal cortex<br />

<strong>of</strong> 14 days MAMAc treated male (A) and female (B) Eker rats, respectively (n=3 per<br />

group). (C-F) BrdU S-Phase mean labeling indices <strong>of</strong> male and female Eker rats<br />

treated with MAMAc for 3 (C-D) and 6 months (E-F), respectively (n=5 per group). Data<br />

represent the mean + SD. Significant differences between treated and time-matched<br />

control groups was analyzed using one-way ANOVA and Bonferroni's Post Test for<br />

multiple comparisons (*p < 0.05; **p < 0.01 and ***p


Chapter 4: Manuscript II<br />

BrdU-immunostaining based analysis <strong>of</strong> cell proliferation demonstrated no significant<br />

changes in cell proliferation rates in proximal tubules <strong>of</strong> three or six month treated male rats<br />

(Figure 4.5, C-F). Female rats showed a transient 1.8 fold increase <strong>of</strong> cell proliferation after<br />

3 months <strong>of</strong> MAMAc treatment in the inner cortex, whereas this was not observable after 6<br />

months <strong>of</strong> treatment (Figure 4.5, D and F). Distal tubules and collecting ducts did not show<br />

a significant increased cell proliferation compared to the control animals (data not shown).<br />

4.4.5 Detection <strong>of</strong> O6MG in MAMAc treated Eker-rats<br />

LC-MS/MS analysis <strong>of</strong> 100µg <strong>of</strong> pooled DNA, from three animals <strong>of</strong> the same treatment<br />

group i.e. 1, 7 and 14 days treatment, demonstrated a time-dependent accumulation <strong>of</strong><br />

O6MG (Figure 4.6). Concurrent O6MG adduct levels in the corresponding control rats were<br />

under the limit <strong>of</strong> detection. MAMAc treated female rats demonstrated slightly higher O6MG<br />

adduct levels than the corresponding treated males at all time-points examined.<br />

Figure 4.6: Number <strong>of</strong> O6MG adducts per 106 dG detected via LC-MS/MS in DNA <strong>of</strong> male and<br />

female Eker rats after 1, 7 and 14 days <strong>of</strong> MAMAc treatment (n=1, with DNA pooled<br />

from three replicate animals)<br />

88


4.5 Discussion<br />

Chapter 4: Manuscript II<br />

Cycasin and its active metabolite MAM(Ac) were shown to be carcinogenic in rodents when<br />

administered at very high doses (Hirono et al. 1968; Laqueur 1968; Fukunishi et al. 1971).<br />

However, the use <strong>of</strong> high doses <strong>of</strong> carcinogens may not reflect the carcinogenic potential <strong>of</strong><br />

a test compound und realistic exposure scenarios, since unspecific additional enhancing<br />

effects such as cytotoxicity and regenerative cell proliferation may occur at the high doses<br />

employed (Ames and Gold 1990; Foran 1997; Cohen and Arnold 2008). In addition, dosedependant<br />

differences in metabolism, as well as in pharmacokinetics <strong>of</strong> a compound, could<br />

also have an influence on carcinogenesis.<br />

Previous studies with rats exposed to low doses <strong>of</strong> DNA alkylating compounds<br />

demonstrated that short-term treatment with genotoxic (renal or hepatic) carcinogens<br />

resulted in a characteristic expression pr<strong>of</strong>ile, including the up-regulation <strong>of</strong> numerous<br />

genes involved in DNA damage response, cell cycle arrest and apoptosis (Ellinger-<br />

Ziegelbauer et al. 2004; van Delft et al. 2004; Ellinger-Ziegelbauer et al. 2005; Stemmer et<br />

al. 2007). Consequently and based on its genotoxic properties, it was assumed that<br />

MAMAc would result in a similar expression pr<strong>of</strong>ile as observed for other DNA alkylating<br />

compounds, even when administered at low doses. However, contrary to expectations<br />

treatment with 250µg/kg BW MAMAc did not result in an up-regulation <strong>of</strong> the expected<br />

genes, i.e. genes involved in DNA damage response, cell cycle arrest and apoptosis (Table<br />

4.1). This lack <strong>of</strong> deregulation observed in the microarray analyses was furthermore<br />

corroborated via quantitative real time PCR. For instance, MGMT, an important protein in<br />

the repair <strong>of</strong> alkylated DNA, was unchanged in male and female rats <strong>of</strong> both treatment<br />

groups, and at any exposure time (Supplementary Figure 4.1).<br />

In contrast to the latter, a time-dependent increase <strong>of</strong> MAMAc induced O6-methylguanine<br />

(O6meG) DNA-adducts in male and female Eker rats was observed (Figure 4.6). The lack<br />

<strong>of</strong> a measurable DNA damage response, as suggested from the data presented here, may<br />

be explained by: a) a time-dependent accumulation <strong>of</strong> O6meG DNA-adducts with a<br />

corresponding DNA damage response, yet changes in the DNA damage response genes<br />

were too small to be detected by gene expression analysis; b) the DNA adducts were not<br />

recognized, thereby leading to the observed lack <strong>of</strong> response at the gene expression level;<br />

and c) the DNA adducts were recognized but not sufficiently repaired with the DNA damage<br />

machinery expressed. The question, whether MAMAc-induced O6meG-adducts were<br />

sufficiently repaired and/or were not sufficiently recognized and therefore not repaired and<br />

thus have the potential to result in the onset <strong>of</strong> cancer could be answered by comparing<br />

89


Chapter 4: Manuscript II<br />

total numbers <strong>of</strong> preneoplastic lesions in 3 and 6 months MAMAc treated and control Eker<br />

rats. Indeed, 3 and 6 months exposure to renal carcinogens were previously demonstrated<br />

to be effective in the identification <strong>of</strong> genotoxic compounds in Eker rats (Walker et al. 1992;<br />

McDorman and Wolf 2002; Morton et al. 2002; Stemmer et al. 2007). Within this timeperiod<br />

genotoxic compounds may cause a second hit mutation in the remaining allele <strong>of</strong> the<br />

Tsc2 tumor suppressor gene. Therefore, a higher total number <strong>of</strong> preneoplastic renal<br />

lesions in chronically treated rats would indicate a genotoxic action <strong>of</strong> MAMAc, and an<br />

insufficient DNA damage response, when compared to vehicle control animals.<br />

However, treatment with MAMAc resulted only in a marginal increase <strong>of</strong> total numbers <strong>of</strong><br />

preneoplastic lesions (bATs and bAHs) at all time-points examined, and statistical<br />

significance was only reached in female rats after 3 months <strong>of</strong> treatment (Figure 4.4). The<br />

latter corresponding to the increased cell proliferation observed at 3 but not 6 months <strong>of</strong><br />

MAMAc treatment in female but not in male rats (Figure 4.5). The concurrent increase <strong>of</strong><br />

cell proliferation and preneoplastic lesions not only points to a critical role <strong>of</strong> cell<br />

proliferation for the formation <strong>of</strong> renal preneoplastic lesions, but also suggests a higher<br />

susceptibility <strong>of</strong> female rats towards MAMAc treatment. The latter is corroborated by the<br />

finding that an increased cell proliferation in female but not in male rats was already<br />

detectable after 14 days <strong>of</strong> MAMAc treatment. Since MAMAc treatment did not induce overt<br />

tubular damage, increased cell proliferation may rather result from a mitogenic mode <strong>of</strong><br />

action, than from a regenerative response.<br />

Above data thus suggest higher susceptibility <strong>of</strong> female rats to MAMAc-induced toxicity<br />

and/or carcinogenicity. Indeed, MAMAc treatment resulted in higher O6meG-adduct levels<br />

in females than in males (Figure 4.6). O6meG-adducts in turn may give rise to guanine to<br />

adenine transitions typically detected as a consequence <strong>of</strong> this adducts type. In<br />

combination with a higher MAMAc-induced cell proliferation, these mutations may be fixed<br />

before a sufficient repair is possible, and may explain the significantly increased number <strong>of</strong><br />

pre-neoplastic lesions in female Eker rats. In contrast in male Eker rats, prolonged<br />

treatment with MAMAc resulted in an increased total number and incidence <strong>of</strong> kidney<br />

tumors (adenomas and carcinomas), but not in a significant increase <strong>of</strong> preneoplastic<br />

lesions (Figure 4.4). Thus, in male rats low doses <strong>of</strong> MAMAc may exert their carcinogenic<br />

action rather by accelerating the progression <strong>of</strong> pre-existing (hereditary) preneoplastic<br />

lesions, than by lesion initiation. However, since MAMAc did not increase overall cell<br />

proliferation in male rats, tumor promotion cannot be explained by a mitogenic effect <strong>of</strong><br />

MAMAc. Another possible mechanism leading to MAMAc tumor promotion is suggested<br />

from the gene expression pr<strong>of</strong>iles <strong>of</strong> male Eker rats treated for 1, 3, 7 and 14 days with<br />

MAMAc. Although a relatively small number <strong>of</strong> genes appeared to be deregulated by<br />

90


Chapter 4: Manuscript II<br />

MAMAc, the most prominent gene cluster with 20% <strong>of</strong> all deregulated genes included<br />

down-regulated genes encoding basic transcription factors, and proteins involved in RNA<br />

splicing, translation and folding. This response is in agreement with previous findings <strong>of</strong> an<br />

early and transient inhibition <strong>of</strong> DNA-, RNA- and protein synthesis in the rat liver within the<br />

first hours <strong>of</strong> MAMAc treatment (Zedeck et al. 1970). More importantly, down-regulated<br />

genes included targets that are known for their tumor suppressing properties, such as the<br />

transcription factor PLAGL1, which encodes a growth suppressor protein frequently deleted<br />

in cancer (Abdollahi 2007), or BHLHB3, a candidate tumor suppressor gene in lung cancer<br />

(Falvella et al. 2008), leading to an overall reduced tumor suppressing environment in the<br />

kidney.<br />

In male rats, MAMAc furthermore resulted in an up-regulation <strong>of</strong> 4 genes involved in TGF-ß<br />

signaling, a pathway which plays an important role in the modulation <strong>of</strong> cellular functions<br />

such as immune function, cell proliferation, cell motility, glycolysis and epithelial-tomesenchymal<br />

transition (Leivonen and Kahari 2007). TGF-ß signaling is further known to<br />

posses janus-properties, i.e. it can function as a tumor suppressor at early stages <strong>of</strong> kidney<br />

cancer but it can also promote tumor progression and metastasis in later stages (Gold<br />

1999; Laping et al. 2007). Early stimulation <strong>of</strong> the TGF-ß pathway in male rats could<br />

therefore still be involved in tumor suppression, but may also result in tumor promotion at<br />

later time-points.<br />

In summary, above findings demonstrate that the combination <strong>of</strong> sensitive methods such as<br />

gene expression pr<strong>of</strong>iling and adduct analysis via LC-MS/MS with established methods e.g.<br />

cell proliferation analysis and histopathology allows to detect earliest carcinogen induced<br />

effects at low dose levels and at time-points when histopathological changes are not yet<br />

readily detectable. Above findings suggests that subchronic and –chronic exposure to low<br />

doses <strong>of</strong> MAMAc results in gender-specific differences in the carcinogenic response. In<br />

female rats the increase in early tumor stages and absence <strong>of</strong> late stages may point to a<br />

tumor initiating role <strong>of</strong> MAMAc, while in male rats, the increase in later tumor stages after 6<br />

months exposure is suggestive <strong>of</strong> a tumor promoting function.<br />

Acknowledgement:<br />

We would like to thank the Federal Ministry <strong>of</strong> Education and Research for funding the<br />

project (BMBF: 0313024), Tanja Lampertsdörfer and Gudrun von Scheven, Alexandra<br />

Heussner and Evelyn O’Brien for skilful assistance during the animal experiment, Kerstin<br />

Albrecht for microarray hybridization, Valeriy Afonin for DNA extraction for LC-MS/MS<br />

analysis and Paul T. Pfluger for critically reading the manuscript.<br />

91


4.6 Supplement<br />

4.6.1 Supplementary experimental procedures: Real time PCR<br />

Chapter 4: Manuscript II<br />

To evaluate gene expression by real time PCR, total RNA was extracted from RNAlater<br />

(Qiagen, Germany) fixed renal cortex <strong>of</strong> 14-days MAMAc treated and control male and<br />

female Eker rats (n=3) using RNEasy Mini Kit (Qiagen, Germany), according to<br />

manufacturer’s instructions. Following DNase treatment, reverse transcriptions were<br />

performed using SuperScript III (Invitrogen) and random primers (Invitrogen). Real-time<br />

PCRs <strong>of</strong> the house keeping gene hypoxanthin phosphoribosyl transferase (HPRT: forward<br />

primer: aagacagcggcaagttgaat; reverse primer: ggctgcctacaggctcatag) and <strong>of</strong> candidate<br />

genes O6-methylguanine–DNA methyltransferase (MGMT: forward primer:<br />

cagtaggaggagcgatgagg; reverse primer: gctggaagactcgaaggatg) were performed on an<br />

Eppendorf Cycler using iQ-SybrGreen Supermix (Biorad). All samples were performed in<br />

technical duplicates. Relative amounts <strong>of</strong> gene-specific mRNAs were calculated via Ct<br />

values, based on a standard curve <strong>of</strong> five points with known amounts <strong>of</strong> template cDNA.<br />

4.6.2 Supplementary Figures<br />

Supplementary figure 4.1: Gene expression analysis <strong>of</strong> MGMT in male and female Eker rats<br />

following 14-days treatment with MAMAc. Data <strong>of</strong> three replicate animals are shown<br />

as mean + SEM. One-way ANOVA with Bonferroni's post test, demonstrated no<br />

significant difference <strong>of</strong> male and female MAMAc treated Eker rats vs. the respective<br />

controls.<br />

92


4.6.3 Supplementary Tables<br />

Chapter 4: Manuscript II<br />

Supplementary table 4.1: Non-neoplastic pathology <strong>of</strong> the renal cortex <strong>of</strong> male Eker and wild type rats treated with MAMAc for 1, 3 7 and 14 days respectively<br />

(n=3 per group). Histopathological changes were ranked from none (0) to severe (4) including intermediate classes. Values are presented as<br />

median ± median absolute deviation. Incidences are given in parenthesis.<br />

Pigment<br />

Deposits<br />

Necrosis<br />

Apoptosis<br />

Karyomegally<br />

Vacuolization<br />

Cell shedding<br />

Proteinaceous<br />

casts<br />

Tubular<br />

dilatation<br />

Calcium casts<br />

Control (male Eker rats) MAMAc (maleEker rats) Control (female Eker rats) MAMAc (femaleEker rats)<br />

d1 d3 d7 d14 d1 d3 d7 d14 d1 d3 d7 d14 d1 d3 d7 d14<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.2<br />

(1/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.2<br />

(1/3)<br />

0.5+0.2<br />

(2/3)<br />

0.5+0.2<br />

(2/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.2<br />

(1/3)<br />

0.0+0.0<br />

(0/3)<br />

0.5+0.2<br />

(2/3)<br />

0.0+0.2<br />

(1/3)<br />

0.0+0.2<br />

(1/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.2<br />

(1/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.2<br />

(1/3)<br />

0.0+0.0<br />

(0/3)<br />

0.5+0.2<br />

(2/3)<br />

0.5+0.0<br />

(3/3)<br />

0.0+0.2<br />

(1/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.2<br />

(1/3)<br />

0.5+0.2<br />

(2/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.2<br />

(1/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.2<br />

(1/3)<br />

0.0+0.0<br />

(0/3)<br />

0.5+0.2<br />

(2/3)<br />

0.5+0.0<br />

(3/3)<br />

0.0+0.2<br />

(1/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.2<br />

(1/3)<br />

0.5+0.2<br />

(2/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.2<br />

(1/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.2<br />

(1/3)<br />

0.0+0.0<br />

(0/3)<br />

0.5+0.2<br />

(2/3)<br />

0.5+0.0<br />

(3/3)<br />

0.0+0.2<br />

(1/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.2<br />

(1/3)<br />

0.5+0.2<br />

(2/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.2<br />

(1/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.2<br />

(1/3)<br />

0.0+0.0<br />

(0/3)<br />

0.5+0.2<br />

(2/3)<br />

0.5+0.0<br />

(3/3)<br />

0.0+0.2<br />

(1/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.2<br />

(1/3)<br />

0.5+0.2<br />

(2/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.5+0.0<br />

(3/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.2<br />

(1/3)<br />

0.5+0.3<br />

(2/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.5+0.2<br />

(3/3)<br />

0.5+0.2<br />

(2/3)<br />

0.0+0.2<br />

(1/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.5+0.2<br />

(3/3)<br />

0.5+0.2<br />

(2/3)<br />

0.0+0.2<br />

(1/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.2<br />

(1/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.5+0.2<br />

(2/3)<br />

0.5+0.2<br />

(2/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.5+0.3<br />

(2/3)<br />

0.5+0.0<br />

(3/3)<br />

0.5+0.2<br />

(2/3)<br />

0.0+0.0<br />

(0/3)<br />

93


Tubular<br />

regeneration<br />

CPN<br />

Inflammation<br />

Chapter 4: Manuscript II<br />

Control (male Eker rats) MAMAc (maleEker rats) Control (female Eker rats) MAMAc (femaleEker rats)<br />

d1 d3 d7 d14 d1 d3 d7 d14 d1 d3 d7 d14 d1 d3 d7 d14<br />

0.0+0.0<br />

(0/3)<br />

0.5+0.2<br />

(2/3)<br />

0.0+0.2<br />

(1/3)<br />

0.0+0.2<br />

(1/3)<br />

0.0+0.2<br />

(1/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.2<br />

(1/3)<br />

0.5+0.2<br />

(2/3)<br />

0.0+0.2<br />

(1/3)<br />

0.0+0.2<br />

(1/3)<br />

0.0+0.2<br />

(0/3)<br />

0.0+0.2<br />

(0/3)<br />

0.0+0.2<br />

(1/3)<br />

0.5+0.2<br />

(2/3)<br />

0.0+0.2<br />

(1/3)<br />

0.0+0.2<br />

(1/3)<br />

0.0+0.2<br />

(0/3)<br />

0.0+0.2<br />

(0/3)<br />

0.0+0.2<br />

(1/3)<br />

0.5+0.2<br />

(2/3)<br />

0.0+0.2<br />

(1/3)<br />

0.0+0.2<br />

(1/3)<br />

0.0+0.2<br />

(0/3)<br />

0.0+0.2<br />

(0/3)<br />

Supplementary table 4.2: Incidence and mean numbers <strong>of</strong> preneoplastic and neoplastic lesions <strong>of</strong> the renal cortex. Mean + SD <strong>of</strong> atypical tubule (AT), aypical<br />

hyperplasia (AH) and neoplastic (adenoma, carcinoma) lesions <strong>of</strong> male and female MAMAc treated Eker and wild type rats and their respective<br />

controls (n=3). Incidences <strong>of</strong> respective lesions in treated or control animals are given in parentheses.<br />

Group Sex AT: Mean + SD AH: Mean + SD Neoplastic lesions: Mean + SD<br />

d1 d3 d7 d14 d1 d3 d7 d14 d1 d3 d7 d14<br />

Control male 1.7 ± 0.9 1.3 ± 0.5 2.7 ± 3.8 1.3 ± 1.2 0.7 ± 0.5 0.0 ± 0.0 0.3 ± 0.6 0.3 ± 0.5 0.3 ± 0.5<br />

(3/3) (3/3) (2/3) (2/3) (2/3) (0/3) (1/3) (1/3)<br />

A<br />

0.0 ± 0.0 0.0 ± 0.0 0.0 ± 0.0<br />

(1/3)<br />

(0/3) (0/3) (0/3)<br />

MAMAc male 1.7 ± 1.2 2.0 ± 1.4 1.0 ± 0.8 1.0 ± 0.8 0.0 ± 0.0 0.0 ± 0.0 0.0 ± 0.0 0.0 ± 0.0 0.3 ± 0.5<br />

(2/3) (3/3) (2/3) (2/3) (0/3) (0/3) (0/3) (0/3)<br />

A<br />

0.0 ± 0.0 0.0 ± 0.0 0.0 ± 0.0<br />

(1/3)<br />

(0/3) (0/3) (0/3)<br />

Control female 3.0 ± 2.2 2.0 ± 1.4 1.7 ± 1.5 2.7 ± 0.9 0.0 ± 0.0 0.7 ± 0.5 0.0 ± 0.0 0.3 ± 0.5 0.0 ± 0.0 0.0 ± 0.0 0.0 ± 0.0 0.0 ± 0.0<br />

(2/3) (3/3) (2/3) (3/3) (0/3) (2/3) (0/3) (1/3) (0/3)<br />

(0/3) (0/3) (0/3)<br />

MAMAc female 2.0 ± 0.8 2.7 ± 3.1 2.7 ± 2.5 5.0 ± 1.4 0.3 ± 0.5 0.0 ± 0.0 0.0 ± 0.0 0.0 ± 0.0 0.0 ± 0.0 0.0 ± 0.0 0.0 ± 0.0 0.3 ± 0.5<br />

(3/3) (2/3) (3/3) (3/3) (1/3) (0/3) (0/3) (0/3) (0/3)<br />

(0/3) (0/3)<br />

A<br />

(1/3)<br />

A C<br />

: Adenoma; : Carcinoma<br />

0.0+0.2<br />

(1/3)<br />

0.5+0.2<br />

(2/3)<br />

0.0+0.2<br />

(1/3)<br />

0.0+0.2<br />

(1/3)<br />

0.0+0.2<br />

(0/3)<br />

0.0+0.2<br />

(0/3)<br />

0.0+0.0<br />

(0/3)<br />

0.5+0.0<br />

(3/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.2<br />

(1/3)<br />

0.5+0.2<br />

(2/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.2<br />

(1/3)<br />

0.5+0.2<br />

(2/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.2<br />

(1/3)<br />

0.5+0.2<br />

(2/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.2<br />

(1/3)<br />

0.5+0.2<br />

(2/3)<br />

0.0+0.0<br />

(0/3)<br />

0.0+0.2<br />

(1/3)<br />

0.5+0.0<br />

(3/3)<br />

0.0+0.0<br />

(0/3)<br />

94


Chapter 5: Manuscript III<br />

Establishment <strong>of</strong> a Protocol for the Analysis <strong>of</strong> Laser<br />

Microdissected Rat <strong>Kidney</strong> Samples on Affymetrix Genechips<br />

Kerstin Stemmer a, 1 , Heidrun Ellinger-Ziegelbauer b, 1 , Kerstin Lotz b , Hans-J. Ahr b and<br />

Daniel R. Dietrich a, *<br />

a<br />

Human and Environmental Toxicology, University <strong>of</strong> Konstanz, Konstanz, Germany<br />

b<br />

<strong>Molecular</strong> and Special Toxicology, Bayer Healthcare AG, Wuppertal, Germany<br />

1<br />

These authors contributed equally to this work.<br />

* Corresponding author: Daniel R. Dietrich: daniel.dietrich@uni-konstanz.de<br />

Published in Toxicology and Applied Pharmacology (2006) Nov 15; 217(1):134-42<br />

5.1 Abstract<br />

Laser microdissection in conjunction with microarray technology allows selective isolation<br />

and analysis <strong>of</strong> specific cell populations, e.g., preneoplastic renal lesions. To date, only<br />

limited information is available on sample preparation and preservation techniques that<br />

result in both optimal histomorphological preservation <strong>of</strong> sections and high-quality RNA for<br />

microarray analysis. Furthermore, amplification <strong>of</strong> minute amounts <strong>of</strong> RNA from<br />

microdissected renal samples allowing analysis with gene chips has only scantily been<br />

addressed to date. The objective <strong>of</strong> this study was therefore to establish a reliable and<br />

reproducible protocol for laser microdissection in conjunction with microarray technology<br />

using kidney tissue from Eker rats p.o. treated for 7 days and 6 months with 10 and 1mg<br />

aristolochic acid/kg bw, respectively. <strong>Kidney</strong> tissues were preserved in RNAlater or snap<br />

frozen. Cryosections were cut and stained with either H&E or cresyl violet for subsequent<br />

morphological and RNA quality assessment and laser microdissection. RNA quality was<br />

comparable in snap frozen and RNAlater-preserved samples, however, the<br />

histomorphological preservation <strong>of</strong> renal sections was much better following<br />

cryopreservation. Moreover, the different staining techniques in combination with sample<br />

processing time at room temperature can have an influence on RNA quality. Different RNA<br />

amplification protocols were shown to have an impact on gene expression pr<strong>of</strong>iles as<br />

95


Chapter 5: Manuscript III<br />

demonstrated with Affymetrix Rat Genome 230_2.0 arrays. Considering all the parameters<br />

analyzed in this study, a protocol for RNA isolation from laser microdissected samples with<br />

subsequent Affymetrix chip hybridization was established that was also successfully<br />

applied to preneoplastic lesions laser microdissected from aristolochic acid-treated rats.<br />

5.2 Introduction<br />

An exponentially increasing number <strong>of</strong> publications indicate that microarray technology is<br />

now an accepted and powerful tool for large-scale gene expression pr<strong>of</strong>iling. However, the<br />

outcome and interpretation <strong>of</strong> microarray studies can be strongly affected by inherent tissue<br />

heterogenicity. For example the kidney represents a tissue <strong>of</strong> high complexity with various<br />

structurally and functionally different compartments. It is therefore very difficult to causally<br />

link changes in gene expression patterns from whole kidney homogenates to cell-typespecific<br />

gene expression, the associated pathways and subsequent physiological effects. In<br />

addition, site-specific pathology-related gene expression changes, e.g., within a distinct<br />

tubular segment or a preneoplastic or neoplastic lesion can be diluted by the gene<br />

expression pr<strong>of</strong>ile <strong>of</strong> other cell types in a whole tissue homogenate and thus not be readily<br />

detectable. Laser-assisted microdissection could be a useful tool to overcome such<br />

obstacles. Two <strong>of</strong> the most common technologies are laser capture microdissection (LCM)<br />

(Emmert-Buck et al. 1996) and laser microdissection and pressure catapulting (LMPC)<br />

(Schutze et al. 1998). Both technologies allow the dissection <strong>of</strong> cell populations, single cells<br />

or even sub-cellular structures from frozen or paraffin-embedded tissues. As the<br />

microdissected structures remain morphologically intact and macromolecules are not<br />

damaged by the laser beam, proteins, DNA or RNA can be isolated for subsequent<br />

analyses. However, the combination <strong>of</strong> laser-assisted microdissection and microarray<br />

technology has to cope with three main challenges: (1) minimizing RNA degradation over<br />

several tissue (section) processing steps, (2) preserving tissue morphology for accurate<br />

histological examination <strong>of</strong> lesions selected for microdissection and (3) maintaining the<br />

proportionality <strong>of</strong> specific RNA species after several rounds <strong>of</strong> RNA amplification, required<br />

to obtain sufficient RNA for chip hybridization. It is important to conserve high-quality RNA,<br />

as degraded RNA can result in detection <strong>of</strong> an altered expression pattern (Spiess et al.<br />

2003). RNA must therefore be protected during all processing steps, e.g., tissue harvesting,<br />

storage, sectioning, staining and fixation and finally during microdissection. Most <strong>of</strong> these<br />

steps are conducted at room temperature. The use <strong>of</strong> the commercially available stabilizing<br />

reagent RNAlater (e.g., from Ambion or Qiagen) could therefore be a promising tool for<br />

96


Chapter 5: Manuscript III<br />

RNA preservation in cryo-fixed sections. As a first step <strong>of</strong> the experiments presented here,<br />

the use <strong>of</strong> RNAlater-preserved to snap frozen tissue with respect to tissue morphology and<br />

RNA quality was compared. Furthermore, the influence <strong>of</strong> long term-storage, repeated use<br />

<strong>of</strong> same cryo-samples and different staining techniques on histomorphological tissue<br />

preservation and RNA quality was determined.<br />

Besides maintaining tissue morphology and high-quality RNA, another challenge to be<br />

dealt with is the low amount <strong>of</strong> RNA obtained from microdissected material. Standard<br />

protocols for microarray analysis are based on starting amounts <strong>of</strong> 1–5μg total RNA. Yet<br />

laser-assisted microdissection yields at best nanograms <strong>of</strong> RNA. To overcome this<br />

obstacle, it is necessary to employ RNA amplification methods to generate the required<br />

microgram amounts <strong>of</strong> RNA. Linear one-cycle mRNA amplification by a T7-based in vitro<br />

transcription (IVT) protocol was first described by Van Gelder et al. (Van Gelder et al. 1990)<br />

and became the standard amplification and labeling protocol for the Affymetrix Chip<br />

technology. A second amplification round can be performed to further increase the RNA<br />

yield. However, a major concern remains as to whether the amplification via IVT maintains<br />

the proportionality <strong>of</strong> RNA species present in the original sample, since it was shown<br />

previously that gene expression ratios were not completely conserved between linearly<br />

amplified and non-amplified material (Baugh et al. 2001; Goley et al. 2004; Nygaard et al.<br />

2005). Consequently, the comparability <strong>of</strong> one- and two-cycle amplification protocols on the<br />

gene expression pr<strong>of</strong>iles <strong>of</strong> rat kidney samples was elucidated and presented here. <strong>Kidney</strong><br />

samples were taken from rats treated with vehicle control or Aristolochic acid (AA), a potent<br />

genotoxic renal toxicant and carcinogen that was previously shown to induce tumors in<br />

kidney, forestomach and urinary bladder <strong>of</strong> the rat (Mengs et al. 1982) as well as renal<br />

toxicity, progradient fibrosis and urothelial tumors in women exposed to AA in Chinese<br />

slimming teas (Vanherweghem et al. 1993; Nortier et al. 2000). Based on the results <strong>of</strong> the<br />

experiments presented here, a protocol for RNA isolation from laser microdissected<br />

samples with subsequent Affymetrix chip hybridization was established that was also<br />

successfully applied to preneoplastic lesions laser microdissected from Aristolochic acidtreated<br />

rats.<br />

97


5.3 Material and Methods<br />

5.3.1 Animal treatment and tissue preparation<br />

Chapter 5: Manuscript III<br />

Heterozygous Tsc2 mutant Eker rats were purchased at 6–8weeks <strong>of</strong> age from the<br />

University <strong>of</strong> Texas MD Anderson <strong>Cancer</strong> Center, Smithville, USA. Upon arrival, rats were<br />

allowed to acclimatize to laboratory conditions for 4 weeks. For the short-term experiment<br />

(7-day treatment) three randomly selected male Eker rats were gavaged daily with 10mg<br />

per kg BW Aristolochic acid (AA) per kg BW dissolved in 0.1M sodium bicarbonate, or with<br />

vehicle (0.1Msodium bicarbonate) alone. At the end <strong>of</strong> the experiment, rats were<br />

anesthetized and then killed via exsanguination subsequent to retrograde perfusion with<br />

PBS. <strong>Kidney</strong>s were collected, cross sectioned to 5-mm slices and either snap frozen with<br />

liquid nitrogen and stored at −80°C or fixed in RNAlater (Qiagen, Hilden, Germany)<br />

according to the manufacturer's instructions. For the latter procedure, kidney slices were<br />

incubated at room temperature for at least 1h, kept at 4°C for up to 1 week, and then<br />

removed from the RNAlater reagent. From these kidney slices, kidney cortex was isolated<br />

and cortex and medulla stored separately in cryovials at −80°C. The RNA <strong>of</strong> the kidney<br />

cortex samples fixed in RNAlater was used for the comparison <strong>of</strong> the one- and two-cycle<br />

protocols shown in Figs. 3–5. In the long-term exposure (6months) five male Eker rats per<br />

group were gavaged on 5 days per week for 6months with 1mg per kg BWAA dissolved in<br />

0.1M sodium bicarbonate and with vehicle (0.1M sodium bicarbonate) respectively.<br />

Following treatment, kidneys were collected as described for the short-term experiment<br />

(above) and then stored using two different protocols: one set <strong>of</strong> kidney cross-sections was<br />

stored in RNAlater as described above, and a second set <strong>of</strong> kidney slices was snap frozen<br />

in liquid nitrogen immediately after harvesting and subsequently stored at −80°C. The<br />

kidney samples <strong>of</strong> these animals were used for results shown in (Figs. 1, 2, and 6) and in<br />

all tables.<br />

5.3.2 Sectioning and staining<br />

Due to the high chaotrophic salt concentration <strong>of</strong> the RNAlater reagent, RNAlater preserved<br />

samples will not freeze in the cryostate and thus were thawed and then washed for 2×5min<br />

or 2×15min in ice-cold RNase-free PBS. For embedding samples were refrozen in the<br />

cryostate at −20°C. To avoid interference <strong>of</strong> frozen tissue embedding reagent OCT<br />

(Cryoblock, medite Medizintechnik GmbH, Burgdorf, Germany) with laser efficiency and<br />

RNA isolation, samples were frozen only on a small drop <strong>of</strong> OCT instead <strong>of</strong> being fully<br />

98


Chapter 5: Manuscript III<br />

embedded in the medium. Both RNAlater- and snap-frozen kidney samples were cut to 10μm-thick<br />

sections on a cryotome. Sections were mounted onto RNase-free membranecovered<br />

slides (PALM Microlaser GmbH, Bernried, Germany) and air-dried on ice for 20 s.<br />

Subsequently, sections were fixed in ice-cold 70% ethanol (RNase-free) for 3min and airdried<br />

for 5 to 10min. For H&E (hematoxylin–eosin) staining, air-dried sections were stained<br />

with RNase-free Mayer's hematoxylin (Sigma Aldrich, Taufkirchen, Germany) for 3min,<br />

rinsed with RNase-free tap water and counterstained with RNase-free eosin Y (Sigma<br />

Aldrich, Taufkirchen, Germany), for 3min. For cresyl violet staining, ethanol-fixed sections<br />

were stained with 1% (w/v) cresyl violet acetate (Sigma Aldrich, Taufkirchen, Germany) in<br />

ACS-grade 100% EtOH for 20s. Excess staining solution was removed by placing the<br />

sections on an absorbent surface. Subsequent to H&E or cresyl violet staining, sections<br />

were dehydrated by shortly dipping the sections in 70% and then 100% ethanol. For further<br />

information, see the customer protocol page <strong>of</strong> the PALM homepage at: www.palmmicrolaser.com.<br />

5.3.3 Laser-assisted microdissection<br />

For laser-assisted microdissection, a PALM laser microdissection and pressure catapulting<br />

(LMPC) system (PALM Microlaser GmbH, Bernried, Germany) was used. Areas <strong>of</strong> variable<br />

size were dissected from H&E-stained whole kidney sections (cresyl violet-stained sections<br />

were not used due to the poor histomorphological preservation <strong>of</strong> the sections, see below).<br />

The microdissected samples were collected in a non-contact and contamination-free<br />

manner, immediately lysed by addition <strong>of</strong> 300μl RLT lysis buffer (RNeasy Micro Kit, Qiagen,<br />

Hilden, Germany) containing 1% <strong>of</strong> β-mercaptoethanol (Sigma Aldrich,Taufkirchen,<br />

Germany) and stored at −80°C. For quality control, total RNA was isolated from additional,<br />

non-dissected and unstained whole sections from each kidney examined.<br />

5.3.4 RNA isolation and quality control<br />

RNA <strong>of</strong> microdissected tissue was isolated using the RNeasy Micro Kit (Qiagen, Hilden,<br />

Germany). RNA from whole kidney cortex immersed in RNAlater was isolated with Qiagen<br />

RNeasy Mini Kits. All isolations were performed according to the manufacturer's<br />

instructions. The quality <strong>of</strong> the total RNA was analyzed with the Agilent 2001 Bioanalyzer<br />

(Agilent Technologies GmbH, Germany) using RNA 6000 Nano or Pico chips. Agilent 2100<br />

expert s<strong>of</strong>tware was used to determine RNA quality, designated as RNA Integrity Number<br />

99


Chapter 5: Manuscript III<br />

(RIN). RIN is calculated from the entire electrophoretic trace by Agilent 2100 expert<br />

s<strong>of</strong>tware and thus represents the relative ratios <strong>of</strong> several RNA regions, e.g., the 5S rRNA<br />

region, the region between the 5S and 18S rRNA and the 18S and 28S rRNA bands.<br />

Based on more than 1000 RNA samples <strong>of</strong> different quality, an algorithm was generated<br />

that describes RNA integrity much more reliably then than the mere ratio <strong>of</strong> the ribosomal<br />

(18S and 28S rRNA) bands. A RIN <strong>of</strong> 7–10 is indicative <strong>of</strong> high RNA quality, whereas a RIN<br />

<strong>of</strong> 1 marks a completely degraded RNA sample (Schroeder et al. 2006). For RNA<br />

concentrations ≥5ng/μl (kidney cortex samples), the RNA quantity was determined using<br />

Ribogreen (<strong>Molecular</strong> Probes, Eugene, USA). For lower RNA concentrations (LMPC<br />

samples) the RNA quantity estimate by the Agilent 2100 expert s<strong>of</strong>tware was utilized. Data<br />

were statistically analyzed using GraphPad Prism 4 s<strong>of</strong>tware with oneway ANOVA followed<br />

by Bonferroni's post test for multiple comparisons or Student's t-test for individual<br />

comparisons.<br />

5.3.5 RNA amplification<br />

For the synthesis <strong>of</strong> biotin-labeled cRNAwith one or two rounds <strong>of</strong> amplification, the<br />

Affymetrix One-cycle and Two-cycle kits were used according to the manufacturer's<br />

instructions, the latter in conjunction with the Megascript T7 kit from Ambion (Austin, USA).<br />

5μg <strong>of</strong> total RNAwas used for the one-cycle protocol, during which the RNA is first reversetranscribed<br />

in the presence <strong>of</strong> an oligo-dT primer coupled with a primer sequence for<br />

bacterial T7-polymerase. Following the synthesis <strong>of</strong> the second cDNA strand, this cDNA<br />

template is then transcribed with T7-polymerase in the presence <strong>of</strong> biotin-labeled<br />

ribonucleotides to yield biotin-labeled antisense cRNA. The concentration, corrected for<br />

input RNA, and the RNA quality was determined based on OD 260/280 measurement and<br />

the Agilent 2100 Bioanalyzer pr<strong>of</strong>ile. Up to 50ng <strong>of</strong> total RNA was employed as starting<br />

material with the two-cycle protocol. After the first round <strong>of</strong> cDNA synthesis as described<br />

above, unlabeled antisense cRNA was synthesized with the Megascript T7 kit from Ambion<br />

(Austin, USA). The cRNA from this first round <strong>of</strong> amplification was quantified after column<br />

purification using Ribogreen. The first strand <strong>of</strong> the second round <strong>of</strong> cDNA synthesis was<br />

primed with random primers, whereas the second strand was primed with the oligo-dT-T7<br />

primer as described above. Finally, biotin-labeled cRNA was synthesized, quantified and<br />

quality controlled as described for the one-cycle kit.<br />

100


5.3.6 Affymetrix chip hybridization and expression pr<strong>of</strong>iling<br />

Chapter 5: Manuscript III<br />

15μg <strong>of</strong> the fragmented biotin-labeled cRNA samples was hybridized on Affymetrix Rat<br />

Genome RAE230_2.0 Genechips according to the manufacturer's instructions.<br />

RAE230_2.0 Genechips contain 31,000 probe sets represented by 11 pairs <strong>of</strong> 25mer<br />

oligonucleotides. Each probe pair consists <strong>of</strong> a perfect match oligo (PM) complementary to<br />

the cRNA target sequence, and a mismatch oligo (MM) with a single base change in the<br />

middle to control for background and nonspecific hybridization. Raw data image files (DAT)<br />

were converted into CEL files by averaging the scan data from the 36 pixels per oligo set.<br />

With GeneData Expressionist Refiner s<strong>of</strong>tware, dark and white spots, gradients and<br />

distortions were detected and corrected using the CEL file data. Using the MAS 5.0<br />

statistical algorithms implemented in the Refiner s<strong>of</strong>tware, the intensities <strong>of</strong> all 11 probe<br />

pairs were condensed to one intensity value per probe set associated with a statistical<br />

detection p-value calculated from the intensity differences <strong>of</strong> the PM and corresponding<br />

MM oligos. This p-value indicates how reliably a transcript is detected. After condensing,<br />

which also includes overall microarray background correction, microarrays were scaled to<br />

an average signal intensity <strong>of</strong> 100 after excluding the highest 2% and lowest 2% <strong>of</strong> the<br />

data.<br />

To select significantly deregulated genes between replicate sample groups representing<br />

kidney samples from short-term Aristolochic acid-treated and vehicle control Eker rats, a ttest<br />

(p≤0.005) combined with a 1.7-fold deregulation cut-<strong>of</strong>f implemented in Genedata<br />

Expressionist Analyst s<strong>of</strong>tware was employed. Genes with an intensity <strong>of</strong> expression


5.4 Results<br />

5.4.1 Tissue fixation and staining<br />

Chapter 5: Manuscript III<br />

I: Influence <strong>of</strong> tissue preservation on histology <strong>of</strong> the sections and on RNA<br />

quality<br />

Degradation <strong>of</strong> RNA can result in detection <strong>of</strong> altered gene expression patterns. Therefore,<br />

the question was asked whether tissue preservation with RNAlater would stabilize RNA in<br />

cryosections and whether the required PBS washing steps would negatively influence RNA<br />

stability. Two RNAlater fixed kidney pieces, each <strong>of</strong> three replicate animals was washed for<br />

2×5min or 2×15min in RNase-free, ice-cold PBS. Subsequently, cryosections were cut and<br />

RNA was isolated from a complete unstained and an H&E-stained section. As control, RNA<br />

was isolated from tissue homogenate <strong>of</strong> a third, unwashed RNAlater fixed kidney piece.<br />

Table 5.1 depicts the RNA quality <strong>of</strong> the samples, displayed as RNA integrity number<br />

(RIN).<br />

Table 5.1: Influence <strong>of</strong> tissue preservation on RNA quality. RNAlater-immersed kidney pieces<br />

from three replicate animals were washed for 2x 5min or 2x 15min in ice-cold PBS<br />

before freezing and cryo-sectioning. RNA was isolated from one unstained or one<br />

H&E-stained section each. Control RNA was isolated directly from RNAlater-fixed,<br />

homogenized kidney cortex. RNA quality is displayed as mean RNA integrity number<br />

(RIN) ± SD. One-way ANOVA and Bonferroni’s post test (p< 0.05(*) and p< 0.01 (**))<br />

suggested the absence <strong>of</strong> significant differences between the samples.<br />

Sample Staining Washing in PBS RIN (mean and SD <strong>of</strong> 3 replicates)<br />

Control N/A N/A 8,8 ± 0,5<br />

Section 1 No 2 x 5min 8,8 ± 0,6<br />

Section 2 H&E 2 x 5min 8,0 ± 0,1<br />

Section 3 No 2 x15min 8,6 ± 0,4<br />

Section 4 H&E 2 x15min 7,6 ± 0,7<br />

The RNA quality is very good after 2×5min or 2×15min <strong>of</strong> washing in ice-cold PBS, with no<br />

significant difference to the control RNA from unwashed RNAlater-fixed tissue. The<br />

comparison with the RNA quality retrieved from snap-frozen tissue (Table 5.2, see below),<br />

RNAlater preservation provided for slightly lower RNA quality.<br />

102


Chapter 5: Manuscript III<br />

Table 5.2: Influence <strong>of</strong> long-term storage on RNA quality (RIN) and yield (ng RNA). RNA was<br />

isolated from one complete section from the kidneys <strong>of</strong> 4 replicate animals. <strong>Kidney</strong><br />

samples were snap-frozen rather than RNAlater immersed. The duration the tissues<br />

were kept frozen at –80°C as <strong>of</strong> initial freeze-storing is shown in the first column.<br />

Duration frozen at –80°C RIN (mean ± SD <strong>of</strong> 4 replicates) RNA ng yield (mean ± SD <strong>of</strong> 4 replicates)<br />

5 months 9,4± 0,4 681.6 ±103.3<br />

12 months 9,2 ± 0,2 493.5 ±135.7<br />

<strong>Kidney</strong> morphology from RNAlater-fixed tissue sections was hardly discernible in unwashed<br />

kidney sections and also in sections washed for 2×5min (Figure 5.1, A). Further washing in<br />

PBS appeared to improve the preservation <strong>of</strong> renal tubular morphological <strong>characteristics</strong> <strong>of</strong><br />

the renal cortical structure (Figure 5.1, B). However, cryosections from RNAlater fixed<br />

tissue never achieved the histomorphological quality <strong>of</strong> cryosections from snap-frozen<br />

tissue even when stored up to 12months at −80°C (Figure 5.1, C). Use <strong>of</strong> snap-frozen<br />

kidneys instead <strong>of</strong> RNAlater-fixed tissue appeared to allow a more precise histological and<br />

pathological examination, especially with regard to the identification <strong>of</strong> tubular structural<br />

detail, and thus to facilitate the structural identification required for LMPC.<br />

Figure 5.1: Histology <strong>of</strong> H&E stained sections. (A) Section from RNAlater-fixed tissue after<br />

2x5min washes in PBS. (B) Section from RNAlater fixed tissue after 2x15min washes<br />

in PBS. (C) Section from snap-frozen tissue, without an RNAlater presoak.<br />

103


II: RNA stability in snap-frozen kidney samples<br />

Chapter 5: Manuscript III<br />

Since long-term storage <strong>of</strong> frozen tissue samples may have a negative impact on<br />

preservation <strong>of</strong> histological features and RNA stability, the influence <strong>of</strong> cryo-storage on<br />

histomorphological <strong>characteristics</strong> and RNA quality was investigated with snap-frozen<br />

tissue stored at −80°C for 5 or 12 months post initial cryopreservation. RNA was isolated<br />

from renal cryosections <strong>of</strong> four independent animals. No significant decrease <strong>of</strong> RNA<br />

quality (RIN) or yield (ng) could be observed in frozen tissue stored up to 12months at<br />

−80°C (Table 5.2). To elucidate whether repeated “thawing” <strong>of</strong> tissue samples from −80°C<br />

to cryostate temperature (−18°C to −20°C) has any influence on RNA stability or tissue<br />

morphology, RNAwas isolated from complete cryosections <strong>of</strong> three replicate animals, that<br />

had been snap frozen, stored at −80°C for 12months, and were then subjected to one or<br />

three cycles <strong>of</strong> “thawing” and refreezing. The mean RIN number <strong>of</strong> the respective samples<br />

only slightly decreased from 9.3±0.2 to 8.5±0.2 after the first and the third freeze–thaw<br />

cycle, respectively, yet still providing for high-quality RNA. Microscopic examination <strong>of</strong><br />

multiple thawed samples demonstrated that tissue morphology was not adversely affected,<br />

thus also allowing pathological examination and histological differentiation (see also Figure<br />

5.1, C and above).<br />

III: Influence <strong>of</strong> the duration <strong>of</strong> section staining procedure on RNA quality<br />

Current working procedures require cryosections to be microdissected at room<br />

temperature. Depending on the number <strong>of</strong> areas that have to be collected from one specific<br />

cryosection, the actual dissection could last for several hours. However, both time and<br />

temperature could be critical factors for the isolation <strong>of</strong> high-quality RNA. RNA yield (ng)<br />

and quality (RIN) may in addition be adversely influenced by different staining methods.<br />

However, for kidney pathology H&E staining is indispensable because it facilitates the<br />

distinction <strong>of</strong> morphological features as well as allows the detection <strong>of</strong> characteristic<br />

basophilic and eosinophilic preneoplastic and neoplastic lesions (Dietrich and Swenberg,<br />

1991). Therefore, the influence <strong>of</strong> H&E staining, temperature and LMPC duration on RNA<br />

yield and quality from H&E-stained cryosections was investigated. In addition, RNA stability<br />

<strong>of</strong> H&E-stained sections were compared with that <strong>of</strong> cresyl violet-stained sections, since<br />

cresyl violet is a recommended staining method for subsequent RNA applications<br />

(customer protocol page at: http://www.palm-microlaser.com). The comparison <strong>of</strong> RNA<br />

immediately retrieved from unstained control sections and H&E-stained sections<br />

demonstrated no significant influence <strong>of</strong> staining on RNA quality (Figure 5.2, 1st and 2nd<br />

bar). Additional handling <strong>of</strong> the sections at room temperature, up to 3 h, tendentially but not<br />

significantly decreased RNA quality. Additional handling time, 6h to overnight, decreased<br />

RNA quality significantly with RIN number decreasing to values below 7.0 (Figure 5.2, bar<br />

104


Chapter 5: Manuscript III<br />

2–6). In contrast cresyl violet staining resulted in a significant decrease <strong>of</strong> the RIN number<br />

directly after staining (Figure 5.2, 1st and 8th bar). However, as <strong>of</strong> then RNA quality<br />

appeared to remain stable over time (Figure 5.2, bar 8–12). Storing the differently stained<br />

sections overnight at −20 °C in general appeared to reduce the rate <strong>of</strong> RNA degradation.<br />

When comparing the RIN numbers <strong>of</strong> the H&E with the cresyl violet stained sections within<br />

the first 3 h <strong>of</strong> handling at room temperature, thus representing a reasonable processing<br />

time, H&E sections provided for a higher RNA quality than the corresponding cresyl violetstained<br />

sections. Therefore, RNA <strong>of</strong> high quality that allows additional RNA processing,<br />

e.g., chip hybridization, can be best obtained from H&E-stained cryosections within 3h post<br />

staining.<br />

Figure 5.2: <strong>Kidney</strong> tissue was frozen immediately after sacrifice and stored at –80°C for 12<br />

months. RNA was isolated from one complete kidney section <strong>of</strong> 3 independent<br />

replicate animals. Sections were stained with Hematoxylin and Eosin (H&E, black<br />

bars) or Cresyl Violet (white bars) and subsequently incubated for 0- 6 hours or over<br />

night (ON) at RT or at -20°C. The control sections (1st bar) were lysed immediately<br />

after sectioning. Significant differences, analyzed by Bonferroni’s multiple<br />

comparison test, are indicated using significance levels <strong>of</strong> p< 0.05(*) and p< 0.001<br />

(**).<br />

5.4.2 RNA quality and yield from pooled microdissected samples<br />

To investigate whether the area <strong>of</strong> pooled microdissected samples had an influence on<br />

RNA quality and yield, areas <strong>of</strong> 1mm 2 and 2mm 2 were laser microdissected from H&Estained<br />

cryosections and analyzed. These analyses as shown in table 5.3 suggested that<br />

total RNA from pooled sections were <strong>of</strong> comparably high quality (RIN >8.0). RNA yields<br />

from both 1mm2 or 2mm2 sections (Table 5.3, also see below) indicated that<br />

105


Chapter 5: Manuscript III<br />

microdissection <strong>of</strong> an approximate area <strong>of</strong> at least 2mm 2 was necessary to achieve the<br />

minimum amount <strong>of</strong> RNA (5ng to 100ng <strong>of</strong> total RNA) required for the two-cycle<br />

amplification protocol. For area comparison, a cross-sectioned glomerulus has an average<br />

area <strong>of</strong> 0.02mm 2 .<br />

Table 5.3: RNA quality and yields from pooled microdissected samples. RNA was isolated from<br />

pooled microdissected samples <strong>of</strong> an overall area <strong>of</strong> 1mm 2 or 2mm 2 .<br />

Pooled area RIN (mean ± SD <strong>of</strong> 3 replicates) Yield ng RNA (mean ± SD <strong>of</strong> 3 replicates)<br />

1mm 2 8,0 ±0,2 3,4 ±0,3<br />

2mm 2 8,3 ±0,4 8,8 ±1,7<br />

5.4.3 RNA amplification and microarray hybridization<br />

I: Influence <strong>of</strong> one-cycle and two-cycle amplification on the expression pr<strong>of</strong>ile<br />

Laser-assisted microdissection only yielded nanograms <strong>of</strong> RNA, thus necessitating global<br />

mRNA amplification for an intended subsequent microarray analysis. Therefore, microarray<br />

experiments with different starting amounts <strong>of</strong> kidney RNA were subjected to one- or twocycle<br />

amplification to investigate the influence <strong>of</strong> one-cycle or two-cycle amplification on the<br />

expression pattern. Figure 5.3 depicts that the RNA amplification factor is highly dependent<br />

on the starting amount <strong>of</strong> RNA. Using 10ng <strong>of</strong> starting RNA, two-cycle amplification led to a<br />

12,000- to 14,000-fold increase in RNA yield. With 50ng <strong>of</strong> starting RNA, the same protocol<br />

resulted in 2500- to 3000-fold RNA yield. One-cycle amplification with 5μg <strong>of</strong> starting RNA<br />

led to a 20- to 30-fold increase <strong>of</strong> the RNA yield. Irrespective <strong>of</strong> the amount <strong>of</strong> starting RNA<br />

or the amplification protocol employed, the RNA yield was comparable (Figure 5.3).<br />

106


Chapter 5: Manuscript III<br />

Figure 5.3: Comparison <strong>of</strong> total RNA yields after either one-cycle (OC) amplification with 5µg <strong>of</strong><br />

starting RNA or two-cycle (TC) amplifications with 10ng or 50ng <strong>of</strong> starting material,<br />

respectively. Data represent mean yield ±SD (n = 6 replicates).<br />

The latter observation was tested with a Bonferroni's multiple comparison test which<br />

suggested that no significant differences can be observed. This further suggests a<br />

saturation <strong>of</strong> the amplification process, an artifact that could lead to preferential<br />

amplification <strong>of</strong> certain genes and consequently in the generation <strong>of</strong> aberrant expression<br />

pr<strong>of</strong>iles. To test this hypothesis, the expression pr<strong>of</strong>iles <strong>of</strong> above samples were analyzed<br />

using Affymetrix Rat Genome RAE230_2.0 arrays. The heat maps in figure 5.4 depict the<br />

ratios <strong>of</strong> gene expression calculated by dividing the expression values in the single AAtreated<br />

replicate samples by the mean expression value <strong>of</strong> the corresponding time-matched<br />

control replicates.<br />

Most <strong>of</strong> the genes selected with both the one-cycle protocol and the two-cycle protocol<br />

starting with 10ng were similarly upregulated after one- and two-cycle amplification. Only<br />

ca. 20% <strong>of</strong> the genes were qualitatively differently expressed in the two protocols, i.e.,<br />

approximately 80% <strong>of</strong> the genes were found to be significantly and similarly deregulated<br />

following one-cycle and two-cycle amplification. Furthermore, a preliminary analysis <strong>of</strong><br />

specific genes that were deregulated following AA treatment demonstrated that the same<br />

main biological pathways were identified in the two amplification protocols thus suggesting<br />

that no preferential/aberrant gene expression is observed due to differing amplification<br />

protocols (Stemmer et al., in preparation).<br />

107


Chapter 5: Manuscript III<br />

Figure 5.4: Expression pr<strong>of</strong>iles <strong>of</strong> differentially expressed genes. Genes that were assigned to be<br />

differentially expressed after one (OC)- or Two (TC)- cycle amplification <strong>of</strong> the<br />

indicated amount <strong>of</strong> RNA from rat kidney after treatment with Aristolochic acid for 7<br />

days were subjected to one-dimensional cluster analysis using Euclidian as distance<br />

metric. The heat maps show ratios <strong>of</strong> gene expression calculated by dividing the<br />

expression values in the single treated replicate samples through the mean<br />

expression value <strong>of</strong> all corresponding time-matched control replicates. The left<br />

diagram depicts the genes commonly selected with the TC- (10ng) and the OC-<br />

protocol, the diagram in the middle those specifically selected with the TC (10ng)<br />

protocol, and the diagram on the right those specifically selected with the OC<br />

protocol. The red color illustrates up-regulated and green down-regulated genes<br />

always in comparison to the corresponding control, as indicated by the color scale.<br />

II: Reproducibility <strong>of</strong> RNA amplification and microarray hybridization<br />

Absolute expression pr<strong>of</strong>iles <strong>of</strong> all genes on a microarray may be influenced by the reagent<br />

lots used. Therefore, hybridization <strong>of</strong> cRNA prepared at different dates with different<br />

reagent lots was carried out at varying dates. This was done with RNA from the short-term<br />

AA-treated and vehicle control samples described above and whose separation in the<br />

three-dimensional space (PCA) is illustrated in figure 5.5.<br />

108


Chapter 5: Manuscript III<br />

Figure 5.5.: Principal components analysis based on the expression <strong>of</strong> all genes <strong>of</strong> the RAE230A<br />

subset (15923 probe sets) on the RAE230_2.0 chip. Black and grey dots depict kidney<br />

samples <strong>of</strong> control and <strong>of</strong> aristolochic acid-treated rats, respectively. Ellipses indicate<br />

samples processed with identical protocols. (A) Different amounts <strong>of</strong> the same RNA<br />

sample subjected to One-cycle (OC) or Two-cycle (TC) amplification. (B) 10 ng <strong>of</strong> the<br />

same RNA sample subjected to TC amplification and chip hybridization at two<br />

different dates (Feb05, Oct05). (C) 10 ng <strong>of</strong> the same RNA sample subjected to TC<br />

amplification at two different dates (Feb05, Oct05), yet hybridized on the same day.<br />

The principle components after the one- and two-cycle protocols separate from each other<br />

only in one dimension, whereas the principle components <strong>of</strong> treated and control samples<br />

separate in another dimensions namely the amplification procedure dimension and the<br />

treatment dimension. The 20% <strong>of</strong> the total deregulated genes observed that differed<br />

between the two amplification protocols may have been responsible for the shift in the<br />

dimension observed in the PCA (Figure 5.5, A). From these observations it can be<br />

concluded that genes deregulated between treated and control samples can be detected<br />

reliably, as reported above, but that microarrays from treated samples must be compared<br />

with control microarrays employing the identical amplification and hybridization protocol.<br />

Preparation <strong>of</strong> cRNA at different dates using separate reagent kits leads to a separation <strong>of</strong><br />

the overall expression pr<strong>of</strong>ile in PCA space (Figure 5.5, B). This appears mainly due to the<br />

separate cRNA synthesis and not as a result <strong>of</strong> microarray hybridization at different dates,<br />

an interpretation that is corroborated by the PCA in figure 5.5, C, for which two separate<br />

cRNA preparations were hybridized on the same day, yielding a very similar dimensional<br />

separation as observed for the PCA in figure 5.5, B. Despite the influence <strong>of</strong> amplification<br />

protocols and possibly hybridization, pr<strong>of</strong>ile separation between treated and control is<br />

always maintained. The conclusion <strong>of</strong> the above findings thus would be that in order to<br />

select differentially expressed genes due to a given treatment, samples <strong>of</strong> the control and<br />

treatment groups must be prepared using the identical protocol and within the same<br />

protocol handling run.<br />

109


5.4.4 Performance <strong>of</strong> the established protocol<br />

Chapter 5: Manuscript III<br />

To test the reliability <strong>of</strong> above protocol RNA was isolated from microdissected samples <strong>of</strong><br />

control and AA exposed male Eker rats (n=3) <strong>of</strong> the 6-month experiment for Affymetrix chip<br />

hybridization. Three different types <strong>of</strong> renal lesions, namely basophilic atypical tubules<br />

(bAT) basophilic hyperplasia (bHA) and chronic progressive nephropathy (CPN), as well as<br />

healthy tissue (HT) <strong>of</strong> the same rats were excised via LMPC. RNA (5ng) was isolated from<br />

each sample type (lesion type or HT) and analyzed on Affymetrix RAE230_2.0 chips (one<br />

chip per lesion type or HT and animal). Amplification factors were comparable for excised<br />

sample types. A box plot, displaying the absolute expression values after scaling to a mean<br />

target expression value <strong>of</strong> 100 per microarray, was used as a quality check <strong>of</strong> the<br />

hybridized chips (Figure 5.6). No outliers or significant variations in chip hybridizations were<br />

found. The data thus demonstrated that the procedure developed generated reproducible<br />

and reliable gene expression determinations.<br />

Figure 5.6. Boxplot analyis <strong>of</strong> Affymetrix genechip data obtained from microdissected renal<br />

samples. 5 ng kidney RNA isolated from various sample types <strong>of</strong> rats (n=3) treated<br />

with AA for 6 months. Sample types: healthy tissue (HT), basophilic atypical tubuli<br />

(bAT), basophilic hyperplasia (bHA), and chronic progressive nephropathy (CPN).<br />

Isolated RNA was subjected to two-cycle amplification and Affymetrix RAE230_2.0<br />

chip hybridization. After scaling to a target intensity <strong>of</strong> 100, the expression intensity<br />

distribution <strong>of</strong> each Affymetrix chip, corresponding to a given sampletype and animal,<br />

was visualized in a boxplot format. The box itself is limited by lower and upper<br />

hinges, which correspond to the 25% and 75% quantiles, respectively.<br />

110


5.5 Discussion<br />

Chapter 5: Manuscript III<br />

Gradual degradation <strong>of</strong> RNA can introduce a variable bias in the quantification and<br />

amplification <strong>of</strong> gene transcripts. This degradation, especially during a protocol<br />

encompassing various processing steps, can severely affect the results <strong>of</strong> downstream<br />

applications, e.g., gene expression pr<strong>of</strong>iling. It is therefore crucial to protect the RNA from<br />

degradation in order to obtain reliable gene expression results. RNA integrity numbers can<br />

be used as standard to define an individual minimum threshold <strong>of</strong> RNA quality. For the<br />

study presented here a RIN number threshold <strong>of</strong> 7.0 was used. This threshold was chosen<br />

based on recommendations by the manufacturer Agilent, who also provides the RINcalculating<br />

s<strong>of</strong>tware (Schroeder et al. 2006). The comparison <strong>of</strong> different preservation and<br />

staining techniques demonstrated that RNA with the required high quality as well as optimal<br />

conservation <strong>of</strong> tissue morphology can be obtained from cryosections <strong>of</strong> snap-frozen tissue<br />

samples that were stained with H&E. The latter findings, however, stand in contrast to<br />

previously published data by Ellis et al. (Ellis et al. 2002) where RNAlater-fixed and PBSwashed<br />

pancreas samples provided reasonably good tissue morphology. Thus caution is<br />

advised as to generalizations <strong>of</strong> fixation and processing protocols, while emphasizing that<br />

protocol optimization procedures, as presented here, are advisable for every animal and<br />

organ prior to embarking on expression pr<strong>of</strong>ile analyses <strong>of</strong> microdissected tissue samples.<br />

From all parameters tested, processing <strong>of</strong> cryosections at room temperature had the most<br />

adverse effect on RNA quality, emphasizing that in order to conserve high-quality RNA<br />

from stained cryosections, 3h <strong>of</strong> processing should not be exceeded. In addition to<br />

maintenance <strong>of</strong> RNA quality, standardization <strong>of</strong> RNA amplification and chip hybridization is<br />

<strong>of</strong> utmost importance. Indeed, optimization <strong>of</strong> amplification protocols also entailed starting<br />

with comparable RNA quantities in order to avoid saturation effects, as demonstrated here.<br />

Due to the tubular structure <strong>of</strong> the kidney, microdissected areas <strong>of</strong> comparable size (mm 2 )<br />

can vary in RNA yields. A minimum area <strong>of</strong> 2mm 2 should be microdissected for reliable<br />

RNA quantification and subsequent two-cycle amplification. In addition, it was<br />

demonstrated that samples should be prepared with an identical protocol during the same<br />

time frame and quality analysis by box plot analysis, to ensure reliable gene expression<br />

data. In conclusion, a protocol for laser-assisted microdissection in conjunction with<br />

Affymetrix microarray technology was established that allows investigation <strong>of</strong> rat kidney<br />

gene expression pr<strong>of</strong>iles with high reproducibility and reliability (Figure 5.6). This protocol is<br />

described in detail in Appendix A.<br />

111


Acknowledgments<br />

Chapter 5: Manuscript III<br />

We would like to thank the Federal Ministry <strong>of</strong> Education and Research (BMBF: 0313024)<br />

for funding the project, T. Lampertsdoerfer and G. von Scheven for their skillful assistance<br />

during the whole animal experiment, E. O'Brien and A.H. Heussner for help with the animal<br />

sacrifice.<br />

5.6 Appendix<br />

Recommended protocol for laser-assisted microdissection in conjunction with Affymetrix<br />

microarray technology<br />

1. Collect kidneys from sacrificed animals and section each kidney into ca. 5mm<br />

replicate slices.<br />

2. Wrap kidney slices into sterile (oven baked-RNAse free) aluminium foil and<br />

immediately snap-freeze samples in liquid nitrogen.<br />

3. Use samples directly or store at −80°C.<br />

4. For preparation <strong>of</strong> cryosections, cool cryostat to −20°C and “thaw” the respective<br />

sample from −80°C to cryostat temperature. It is possible to refreeze the samples at<br />

−80°C for later use. However 3 “freeze and thaw” cycles should not be exceeded.<br />

5. Freeze samples onto a small drop <strong>of</strong> OCT embedding media in the cryostat.<br />

6. Prepare 10 μm cryosections and mount onto special RNase-free membrane<br />

covered slides, recommended for the respective microdissection system, e.g., the<br />

LMPC system from PALM Microlaser GmbH, Bernried Germany.<br />

7. Air dry freshly cut sections for 20s in the cryostat and subsequently fix them in<br />

−20°C cold ethanol (70% in RNase-free ddH2O) for 3min.<br />

8. Air dry sections for 10min at RT.<br />

9. Stain sections with RNase-free, ice-cold Mayer's hematoxylin for 3min.<br />

10. Rinse with RNase-free tap water for 3min.<br />

11. Counterstain with RNase-free, ice-cold eosin Y for 3min.<br />

12. Remove excess staining solution on absorbent surface and dehydrate section by<br />

shortly dipping into ice-cold 70% and 100% ethanol.<br />

13. Air-dry sections at RT and directly use for laser-assisted microdissection<br />

employing, e.g., the PALM LMPC system (PALM Microlaser GmbH, Bernried<br />

Germany).<br />

112


Chapter 5: Manuscript III<br />

14. For RNA isolation from microdissected kidney samples pool a minimum area <strong>of</strong><br />

2mm 2 and collect samples into an appropriate sampling cup <strong>of</strong> the respective<br />

microdissection system.<br />

15. After completion <strong>of</strong> the sample collection, immediately lyse pooled samples via<br />

addition <strong>of</strong> 300μl lysis buffer containing 1% β-mercaptoethanol (e.g., component <strong>of</strong><br />

the RNeasy Micro Kit, Qiagen, Hilden, Germany).<br />

16. Do not exceed 3h between staining and lysis <strong>of</strong> pooled samples.<br />

17. Isolate RNA using e.g. RNeasyMicro Kit (Qiagen, Hilden, Germany) according to<br />

the manufacturer's instruction.<br />

18. Analyze total RNA quality and estimate quantity, using an Agilent Bioanalyzer. For<br />

later amplification and hybridization procedures, use only RNA samples with a RIN<br />

numbers >7.0.<br />

19. Amplify RNA and convert to biotin-labeled cRNA using e.g. the Affymetrix Twocycle<br />

Target labeling kit according to the manufacturer’s instructions. Always use<br />

same starting quantity <strong>of</strong> RNA, e.g., 5ng or 10ng RNA.<br />

20. Use 15μg biotin-labeled cRNA samples for Affymetrix chip hybridization and<br />

process the hybridizations according to the manufacturer's instruction.<br />

113


Chapter 6: Manuscript IV<br />

Earliest preneoplastic renal lesions provide an in-depth<br />

understanding <strong>of</strong> renal carcinogenesis<br />

Kerstin Stemmer 1 , Heidrun Ellinger-Ziegelbauer 2 , Hans-J. Ahr 2 and Daniel R. Dietrich 1 *<br />

1<br />

Human and Environmental Toxicology, University <strong>of</strong> Konstanz, Konstanz, Germany<br />

2<br />

<strong>Molecular</strong> and Special Toxicology, Bayer Healthcare AG, Wuppertal, Germany<br />

* Corresponding author: Daniel R. Dietrich: daniel.dietrich@uni-konstanz.de<br />

American Journal <strong>of</strong> Pathology, submitted<br />

6.1 Abstract<br />

Although the mTOR pathway appears critical both in human and rat renal tumors, it<br />

remains to be established whether both TORC1 and TORC2 pathways are involved in the<br />

genesis and/or progression <strong>of</strong> renal preneoplastic lesions to tumors. Eker rats<br />

heterozygous for a mutation in the TSC2 tumor suppressor gene, thus predisposed for the<br />

early development <strong>of</strong> renal lesions with morphologic similarities with human renal cancer,<br />

were used to analyze the specific involvement <strong>of</strong> TORC1 and TORC2 in the carcinogenand<br />

sex-dependent formation and progression <strong>of</strong> preneoplastic lesions.<br />

Our data demonstrate activation <strong>of</strong> both, TORC1 and TORC2 dependent mTOR pathways<br />

and comparable gene expression pr<strong>of</strong>iles in laser-microdissected preneoplastic lesions<br />

from genotoxic (aristolochic acid) and non-genotoxic (ochratoxin A) treated and control<br />

Eker rats. Co-localised immunostaining <strong>of</strong> phospho-ribosomal protein S6, phospho-Akt<br />

(Ser473) and cytoplasmatic FOXO1 confirmed TORC1 and TORC2 activation, thus<br />

corroborating disruption <strong>of</strong> the mTOR-AKT feedback inhibition in preneoplastic lesions.<br />

Moreover, the here presented findings strongly suggest that the compound-dependent<br />

formation <strong>of</strong> preneoplastic lesions is limited to a critical period <strong>of</strong> time, while lesion<br />

progression appears compound-independent. Therefore, preneoplastic lesion initiation<br />

seems to be influenced by multiple events, whereas clonal expansion appears primarily<br />

dependent on the deregulation <strong>of</strong> the AKT-TSC2-TORC1 and TORC2-AKT pathways.<br />

114


Chapter 6: Manuscript IV<br />

Together these findings indicate that continuous dosing with carcinogens may not have an<br />

augmenting influence exciting lesions. In addition, further in-depth understanding <strong>of</strong> the<br />

TORC1 and TORC2 balance in preneoplastic lesions may be critical for improved renal<br />

cancer treatment strategies.<br />

6.2 Introduction<br />

The mammalian target <strong>of</strong> rapamycin (mTOR) pathway is upregulated in several human<br />

cancers (Albanell et al. 2007), amongst which are renal cell carcinomas (Pantuck et al.<br />

2006; Radulovic and Bjelogrlic 2007). Under non-pathological conditions this pathway is<br />

tightly controlled by the tumor suppressor complex tuberous sclerosis 2 (TSC2, tuberin)<br />

and its obligate binding partner, TSC1 (hamartin) (Jozwiak 2006). Inactivating mutations <strong>of</strong><br />

TSC2 were shown to be accompanied by Rheb mediated activation <strong>of</strong> the raptor containing<br />

mTOR complex 1 (TORC1) and its effectors p70S6K, S6 ribosomal protein (S6RP), 4E-<br />

BP1 and eIF4G (Kenerson et al. 2002; Mak and Yeung 2004). As a consequence, TORC1<br />

dependent protein translation is activated, affecting several cell growth regulators, e.g.<br />

stabilization <strong>of</strong> the transcription factor hypoxia-inducible-growth-factor α (HIF1α)<br />

(Brugarolas et al. 2003), which in turn can drive diverse processes involved in<br />

tumorigenesis, including cell growth, angiogenesis and glycolysis (Semenza 2002).<br />

Moreover, the antiproliferative capacity <strong>of</strong> TSC2 was shown to be inhibited by the c-Myc<br />

oncogene in vitro (Rosner et al. 2003; Ravitz et al. 2007), demonstrating the broad<br />

dimension <strong>of</strong> the TSC2 tumor suppressor network. To date, it remains to be established,<br />

whether TSC2 is also involved in the regulation <strong>of</strong> rictor containing mTOR complex 2<br />

(TORC2). This complex was previously shown to function as an important regulator <strong>of</strong> the<br />

cytoskeleton through members <strong>of</strong> the Rho small GTPase family and phosphorylation <strong>of</strong><br />

Protein Kinase Cα (PKCα) (Jacinto et al. 2004; Sarbassov et al. 2004), and to<br />

phosphorylate AKT at Ser473, one <strong>of</strong> the key residues, required for full AKT activation<br />

(Sarbassov et al. 2005). AKT regulates numerous signal transduction processes involved in<br />

cell survival and proliferation, one being direct phosphorylation and inactivation <strong>of</strong> tumor<br />

suppressor FOXO1 (Manning and Cantley 2007).<br />

Although the mTOR pathway was described to be altered in human renal cancer (Pantuck<br />

et al. 2006) and functional loss <strong>of</strong> TSC2 was found in spontaneous and chemically-induced<br />

rat renal tumors (Walker et al. 1992; Kubo et al. 1994; Kenerson et al. 2002), it remains to<br />

be established whether the TORC1 and TORC2 pathways are involved in the formation<br />

115


Chapter 6: Manuscript IV<br />

and/or progression <strong>of</strong> renal preneoplastic lesions to tumors. Furthermore the influence <strong>of</strong><br />

genotoxic and non-genotoxic carcinogens on these pathways remains unknown.<br />

To characterize the specific involvement <strong>of</strong> TORC1 and TORC2 in the carcinogen- and<br />

sex-dependent formation and progression <strong>of</strong> preneoplastic lesions, Eker rats carrying a<br />

heterozygous loss-<strong>of</strong>-function mutation in the TSC2 gene, therefore predisposed for the<br />

early development <strong>of</strong> renal lesions with morphologic similarities with human renal cancer<br />

(Yeung et al. 1995; Hino 2004), appeared an ideal model. Therefore, male and female Eker<br />

rats were treated for 3 and 6 months with low doses <strong>of</strong> the genotoxic aristolochic acid (AA)<br />

(Mengs et al. 1982; Arlt et al. 2002) and the non-genotoxic carcinogen ochratoxin A (OTA),<br />

for which a 10-fold higher tumor incidence was found in male than in female F344 rats<br />

(Boorman et al. 1992; O'Brien and Dietrich 2005). Both are associated with the occurrence<br />

<strong>of</strong> human urothelial tumors and nephropathy in Balkan countries, suggesting a direct link<br />

between human and rat renal carcinogenesis (Stoev 1998; Grollman et al. 2007).<br />

Compound-induced non-neoplastic and neoplastic renal pathology, site-specific renal cell<br />

proliferation, incidence, phenotype and progression stage <strong>of</strong> the preneoplastic lesions, and<br />

the rate <strong>of</strong> preneoplastic lesion progression were determined at the 3- and 6-month timepoint<br />

in both sexes. For the first time, laser-microdissected preneoplastic lesions and<br />

healthy tubules <strong>of</strong> AA and OTA treated as well as control male Eker rats were analyzed<br />

using microarrays, allowing to investigate gene expression pr<strong>of</strong>iles specifically induced by<br />

AA and OTA and to differentiate pathways specific for the progression stage <strong>of</strong><br />

preneoplastic lesions. Activation <strong>of</strong> TORC1 and TORC2 pathways were visualized via<br />

immunohistochemical detection <strong>of</strong> phosphorylated downstream targets.<br />

6.3 Material and Methods<br />

6.3.1 Compounds<br />

Aristolochic acid sodium salt mixture (AA I: 41% and AA II: 56%) was purchased from<br />

Sigma Aldrich, Germany. Ochratoxin A (>98% purity) was kindly provided by M.E. Stack,<br />

US FDA, Washington DC.<br />

6.3.2 Animals<br />

Eker rats were purchased at 6-8 weeks <strong>of</strong> age from the University <strong>of</strong> Texas MD Anderson<br />

<strong>Cancer</strong> Center, Smithville, USA. Groups <strong>of</strong> females and males were randomly assigned to<br />

116


Chapter 6: Manuscript IV<br />

dose groups (10 rats per compound (or vehicle) and time-point) and acclimatized for 4<br />

weeks. Groups <strong>of</strong> 10 Eker rats per sex were gavaged with OTA (210µg/kg BW) or AA<br />

(1mg/kg BW), dissolved in 0.1M NaHCO3 at five days a week. Time-matched vehicle<br />

controls were gavaged with 0.1M NaHCO3. 5 <strong>of</strong> 10 rats per dose group and sex were s.c.<br />

implanted with osmotic ALZET-pumps (Model 2ML1, Charles River Laboratories, Germany)<br />

containing 5-bromo-2-deoxyuridine (BrdU, 20mg/ml sterile saline, Sigma Aldrich, Germany)<br />

5 days prior to sacrifice to allow post-mortem immunohistochemical analysis <strong>of</strong> cell<br />

proliferation. After 3 and 6 months <strong>of</strong> treatment, anesthetized rats were sacrificed by<br />

exsanguination subsequent to retrograde perfusion with PBS and kidneys were collected<br />

(Supplementary figure 6.1).<br />

6.3.3 Sample collection<br />

One half <strong>of</strong> the freshly isolated kidney was cross-sectioned to 5mm slices, slices snap<br />

frozen in liquid nitrogen and stored at -80°C for subsequent cryosectioning. The other half<br />

was fixed in PBS buffered fixative (2% paraformaldehyde and 1% glutaraldehyde) for<br />

subsequent paraffin embedding and sectioning.<br />

6.3.4 Histopathology<br />

H&E stained sections from 10 rats per group were randomized for histopathological<br />

evaluation. Non-neoplastic pathology was classified as none (0), mild (1), moderate (2),<br />

strong (3), and severe (4), while preneoplastic and neoplastic lesions were classified<br />

according to the lesion type (Dietrich and Swenberg 1991) and numbers <strong>of</strong> each lesion type<br />

counted.<br />

6.3.5 Immunohistochemistry<br />

Cell proliferation was evaluated via BrdU-immunohistochemistry. Immunostaining was<br />

carried out as described previously (Stemmer et al. 2007), yet using a monoclonal mouse<br />

anti-BrdU primary antibody (MU247-UC, Biogenex, USA) diluted 1:100 in Power BlockTM<br />

(BioGenex, USA) in an overnight application at 4°C. Cell proliferation was quantified on<br />

randomized sections. 20 microscopic fields (10x ocular, 40x objective) were randomly<br />

chosen within the area <strong>of</strong> the renal outer cortex and inner cortex/outer medulla. Proximal<br />

and distal tubules and collecting ducts were counted separately per field, distinguishing<br />

117


Chapter 6: Manuscript IV<br />

between negative and positive BrdU-stained nuclei. Nuclear labeling indices for BrdU (LI%)<br />

(BrdU positive nuclei/total number <strong>of</strong> nuclei counted) were determined. Subsequently, the<br />

LIs <strong>of</strong> proximal tubules without pathological changes were determined. For LI(%)determination,<br />

at least 1.000 and 500 nuclei were counted in proximal and distal tubules<br />

and collecting ducts, respectively.<br />

Immunohistochemistry <strong>of</strong> phospho-S6 ribosomal protein (pS6RP), phospho-Akt (pAKT) and<br />

FOXO1 in paraffin sections was carried out according to manufacturers’ instructions (Cell<br />

Signaling Technology). Primary rabbit anti-pS6RP (Ser235/236), (Cat No: 2211) was<br />

diluted 1:100, while rabbit anti-p-AKT (Ser473) (Cat No: 3787), was diluted 1:10 and rabbit<br />

anti-FOXO1 (Abcam ab39656) 1:200 in 5% normal goat serum. Antigen–antibody<br />

complexes were visualized using the Super SensitiveTM (BioGenex, USA) alkaline<br />

phosphatase-labeled, biotin–streptavidin amplified detection system and Fast Red as<br />

chromogen.<br />

6.3.6 Laser microdissection and RNA isolation<br />

For microdissection <strong>of</strong> preneoplastic lesions from H&E stained renal cryosections, a laser<br />

microdissection and pressure catapulting (LMPC) system (ZEISS, Germany) was used.<br />

Atypical tubule (bAT), basophilic atypical hyperplasia (bAH) or healthy tubules (HT) were<br />

micodissected separately (Stemmer et al. 2006) from each <strong>of</strong> three replicate rats per dose<br />

group. bAT, bAH and HT from each individual animal were pooled. RNA isolation from<br />

pooled samples and subsequent Affymetrix Rat Genome RAE_230A_2.0 chip hybridization<br />

was carried out as previously described (Stemmer et al. 2006).<br />

6.3.7 Microarray data processing and analysis<br />

Microarray quality control was performed as described previously (Ellinger-Ziegelbauer et<br />

al. 2005) and gene expression data were submitted to the NCBI-GEO repository (GSE<br />

10608). For statistical analysis Expressionist Analyst s<strong>of</strong>tware (Genedata AG, Switzerland)<br />

was used. Significantly deregulated genes per compound and lesion type vs. healthy<br />

tubules <strong>of</strong> control rats were selected by student’s T-test with a p-value cut-<strong>of</strong>f <strong>of</strong> 0.005,<br />

combined with a 2.0-fold deregulation threshold. Identical cut-<strong>of</strong>fs were applied for<br />

comparing healthy tubules <strong>of</strong> AA and OTA treated rats with healthy tubules <strong>of</strong> control rats.<br />

Heatmaps were used to graphically display the relative expression data after one-<br />

118


Chapter 6: Manuscript IV<br />

dimensional clustering <strong>of</strong> the genes. See supplemental information for details on the<br />

validation <strong>of</strong> microarray data.<br />

For functional analysis, significantly deregulated genes were characterized according to the<br />

biochemical role <strong>of</strong> its encoded protein, using information from databases, e.g. NetAffx,<br />

Swissprot, Proteome and Pubmed. Depending on the direction <strong>of</strong> deregulation, genes were<br />

further assigned to pathophysiological categories allowing the comparison <strong>of</strong> deregulated<br />

pathways in different types <strong>of</strong> lesions and healthy tubules.<br />

6.3.8 Statistical analysis<br />

Statistical analysis <strong>of</strong> histopathological and cell proliferation data were carried out using<br />

GraphPad Prism 4® (USA). Significant differences in nuclear labeling indices or total<br />

number <strong>of</strong> lesions in treated and control rats were analyzed by Bonferroni and Dunnett’s<br />

test for multiple comparisons. Lesion incidences were tested for significance using the 2sided<br />

Fisher’s exact test. Ranked non-neoplastic pathology data were analyzed using the<br />

nonparametric Mann-Whitney test.<br />

6.4 Results<br />

6.4.1 Non-neoplastic pathology<br />

Treatment <strong>of</strong> male and female Eker rats with AA or OTA resulted in increased nonneoplastic<br />

renal pathology compared to respective control rats (Supplementary table 6.1).<br />

OTA treatment significantly and time-dependently increased chronic progressive<br />

nephropathy (CPN), as characterized by a thickened basal membrane, an increased<br />

regeneration and monocytes infiltration in both sexes (Supplementary figure 6.2). In<br />

agreement with previously described OTA-induced non-neoplastic pathology (Boorman et<br />

al. 1992; Rasonyi et al. 1999), male and female Eker rats treated with OTA for 6 months<br />

presented with increased apoptosis, necrosis, karyomegally, prevalence <strong>of</strong> dilated tubule<br />

and protein casts, as well as an increased tubular regeneration and inflammatory response<br />

primarily within the inner cortex. In contrast, treatment with AA resulted in weaker pathology<br />

compared to OTA treated rats. A significantly enhanced severity <strong>of</strong> CPN and inflammation<br />

and an increased prevalence <strong>of</strong> dilated tubule and protein casts in male rats were observed<br />

only at the 6-month time-point. Moreover, AA treated females generally presented with<br />

weaker pathology than males.<br />

119


6.4.2 Cell proliferation<br />

Chapter 6: Manuscript IV<br />

Site-specific cell proliferation (Supplementary figure 6.2, C-F), i.e. specific to proximal<br />

tubule, distal tubule or collecting ducts, was assessed via BrdU immunostaining (Table<br />

6.1). Female Eker rats at the 6-month time-point showed an approximately 2-3-fold higher<br />

basal cell proliferation rate than males at all sites, whereas this was not apparent at the 3month<br />

time-point.<br />

Table 6.1: Comparison <strong>of</strong> BrdU S-Phase labeling indices (LI%) <strong>of</strong> Eker rats treated with AA or<br />

OTA for 3 and 6 months, respectively. LI%, determined for proximal tubules (PT)<br />

within randomly chosen fields <strong>of</strong> the outer or inner cortex, <strong>of</strong> healthy proximal tubule<br />

(hPT), distal tubule (DT) and collecting ducts (CD). For PT >1000 nuclei were counted,<br />

for DT and CD >500 nuclei were counted. LI% were tested for significance using oneway<br />

ANOVA and Bonferroni's Post Test for multiple comparisons.<br />

Male Eker rats Female Eker rats<br />

3 months treatment Control AA OTA Control AA OTA<br />

# animals 5 5 5 5 5 5<br />

PT outer cortex 2.4 + 0.9 2.1 + 0.7 1.9 + 0.8 1.8 + 0.4 3.3 + 2.1 2.5 + 0.8<br />

PT inner cortex 2.0 + 1.0 2.2 + 1.0 12.3 + 3.5 b,d 2.2 + 0.3 4.8 + 1.5 12.6 + 4.0 b,d<br />

Male Eker rats Female Eker rats<br />

6 months treatment Control AA OTA Control AA OTA<br />

# animals 5 5 5 5 3 5<br />

PT outer cortex 2.0 + 0.4 4.1 + 2.1 4.9 + 0.7 3.7 + 1.2 6.7 + 0.6 4.8 + 0.8<br />

PT inner cortex 2.7 + 1.3 6.6 + 2.8 a 15.6 + 2.0 b,d 4.9 + 1.8 12.2 + 0.8 b,c,e 14.8 + 3.3 b,d<br />

hPT, outer cortex 1.9 + 0.4 2.3 + 0.6 2.6 + 0.6 4.4 + 0.7 5.3 + 0.3 3.9 + 0.6<br />

hPT, inner cortex 1.9 + 0.5 1.9 + 0.4 2.8 + 0.9 4.9 + 1.1 6.3 + 1.3 4.9 + 1.9<br />

DT 1.9 + 1.1 0.9 + 0.5 1.2 + 0.4 4.5 + 2.5 5.3 + 0.8 4.2 + 1.5<br />

CD 1.5 + 1.7 0.9 + 0.5 1.9 + 2.0 4.4 + 1.9 5.4 + 0.2 5.8 + 2.0<br />

a b<br />

Significantly higher than the respective control group (p< 0.05), Significantly higher than the respective<br />

control group (p< 0.001), c Significantly higher in inner, than outer cortex, within the same treatment group<br />

(p< 0.05), d Significantly higher in inner, than outer cortex, within the same treatment group (p< 0.001), e<br />

Significantly higher in 6 months than 3 months treated rats (same sex, compound and tubular segment)<br />

(p< 0.001)<br />

OTA: 3 months OTA-treatment significantly increased cell proliferation rates 6.2-fold and<br />

5.7-fold above controls in the proximal tubules <strong>of</strong> the inner cortex <strong>of</strong> male and female rats,<br />

respectively. In contrast, a higher cell proliferation was observed following 6 months OTAtreatment<br />

in the inner cortex <strong>of</strong> male rats (5.8-fold) compared to females (3.0-fold).<br />

120


Chapter 6: Manuscript IV<br />

AA: 3 months AA-treatment did not change cell proliferation rates in the proximal tubules <strong>of</strong><br />

the inner or the outer cortex in both male and female rats. However, at the 6-month timepoint<br />

a 2.4- and 2.5-fold increase in cell proliferation in the proximal tubules <strong>of</strong> the inner<br />

cortex was observed in both males and females, respectively (Table 6.1).<br />

When restricting cell proliferation analysis to proximal tubules with a normal (nonpathologic)<br />

phenotype (Supplementary figure 6.2, A), mean cell proliferation rates were not<br />

significantly different in the outer or inner cortex <strong>of</strong> control and treated rats <strong>of</strong> both sexes<br />

(Table 6.1). No increased cell proliferation was observed in proximal tubules <strong>of</strong> the outer<br />

cortex, distal tubule or collecting ducts <strong>of</strong> either sex, irrespective <strong>of</strong> the treatment.<br />

6.4.3 (Pre-) neoplastic pathology<br />

Preneoplastic (atypical tubule (AT) and atypical hyperplasia (AH)) and neoplastic (adenoma<br />

and carcinoma) lesions observed in male and female Eker rats after 3 or 6 months <strong>of</strong><br />

treatment were primarily <strong>of</strong> the basophilic phenotype i.e. bAT (Figure 6.1, A) and bAH<br />

(Figure 6.1,B). Other lesion types, e.g. oncocytic tubules and cysts, were rare and thus not<br />

evaluated further. Despite a 100% incidence <strong>of</strong> bATs in all treatment and control groups<br />

(Figure 6.1, C and D, Supplementary table 6.2), a time- and compound-dependent increase<br />

<strong>of</strong> total numbers <strong>of</strong> bATs and bAHs was observable in treated males and females, when<br />

compared to the respective controls at both time points.<br />

OTA: 3 months OTA-treatment induced a 3.3-fold and a 2.8-fold increase in total bATs in<br />

male and female rats, respectively. A similar increase in bATs was also observed at the 6month<br />

time-point, when compared to the respective controls, suggesting continued OTAinduced<br />

bAT formation in all groups. In males, total numbers <strong>of</strong> bAH increased approx. 3fold<br />

over the time, while the total number <strong>of</strong> bAH in OTA-treated females remained at<br />

similar levels at both time-points, indicating a higher formation <strong>of</strong> preneoplastic lesions in<br />

male rats. This was confirmed by the 80% bAH incidence in male rats compared to the<br />

30% incidence in females (Figure 6.1, D).<br />

AA: A significant 3.0- and 5.6-fold increase <strong>of</strong> the bATs/rat was observed in female rats at<br />

the 3-and 6-month time-points, while male Eker rats showed a significant 3.5 fold increase<br />

<strong>of</strong> bATs at the 6-month time-point only (Figure 6.1, D). Similarly, both male and female rats<br />

presented with a higher total number <strong>of</strong> bAH following 3 and 6-months AA-treatment when<br />

compared to the corresponding controls. However, the bAH/rat ratio was significantly<br />

higher at the 6-month time-point only in female rats.<br />

121


Chapter 6: Manuscript IV<br />

Figure 6.1. Representative preneoplastic lesions as observed in H&E stained renal sections (A) bAT; (B) bAH. Mean number <strong>of</strong> preneoplastic lesions in<br />

AA- and OTA-treated male and female control Eker rats after 3 (C) and 6 months (D). Grey bars: bAT; white bars: bAH. Dunnett’s test: *p < 0.05;<br />

**p < 0.01.<br />

122


6.4.4 Gene expression pr<strong>of</strong>iling<br />

Chapter 6: Manuscript IV<br />

As preneoplastic lesions exhibited a similar basophilic phenotype in both sexes, gene<br />

expression pr<strong>of</strong>ile analyses were restricted to preneoplastic lesions <strong>of</strong> male Eker rats.<br />

Laser microdissected preneoplastic lesions (bAT and bAH) and healthy tubules <strong>of</strong> AA- and<br />

OTA-treated and control males were analyzed using microarrays in order to investigate<br />

gene expression pr<strong>of</strong>iles induced by AA and OTA as well as to differentiate pathways<br />

specific for the progression stage <strong>of</strong> the preneoplastic lesions.<br />

Preneoplastic lesions: Gene expression pr<strong>of</strong>iling <strong>of</strong> microdissected lesions resulted in<br />

570 significantly deregulated annotated and non-redundant genes when compared to<br />

healthy tubules <strong>of</strong> control rats. Although different total numbers <strong>of</strong> genes appeared to be<br />

significantly deregulated in different samples (Table 6.2), visualization <strong>of</strong> expression<br />

pr<strong>of</strong>iles <strong>of</strong> the union <strong>of</strong> selected genes revealed a qualitatively similar expression pr<strong>of</strong>ile in<br />

all preneoplastic lesions, irrespective <strong>of</strong> treatment or lesion type (Figure 6.2).<br />

Figure 6.2: Heatmap comparing gene expression changes <strong>of</strong> microdissected bATs and bAHs <strong>of</strong><br />

AA, OTA and vehicle treated Eker rats, and microdissected healthy tubule from AA<br />

and OTA treated rats. Expression pr<strong>of</strong>iles were compared to microdissected healthy<br />

tubules (HT) <strong>of</strong> control rats. Gene expression ratios are indicated by the color scale:<br />

Red: upregulated; Green: downregulated.<br />

123


Chapter 6: Manuscript IV<br />

Healthy tubules (HT): Gene expression pr<strong>of</strong>iles <strong>of</strong> microdissected morphologically<br />

“healthy” tubules (Supplementary figure 6.2, A) <strong>of</strong> AA- and OTA-treated rats were<br />

compared with HT <strong>of</strong> the respective control rats HT(C) to evaluate the effect <strong>of</strong> AA and<br />

OTA treatment on gene expression in HT after 6 months treatment. Seven and six genes<br />

were significantly deregulated in HT <strong>of</strong> AA and OTA treated males, respectively <strong>of</strong> which<br />

the most important ones are listed (Supplementary table 6.3).<br />

Functional analysis <strong>of</strong> significantly deregulated genes: Functional analysis <strong>of</strong> genes<br />

deregulated in preneoplastic lesions <strong>of</strong> treated and control rats identified a number <strong>of</strong><br />

pathophysiologically relevant endpoints. Table 6.2 specifies the criteria used to assign<br />

deregulated genes to these categories. The most prominent categories, containing the<br />

majority <strong>of</strong> deregulated genes, were similar in all lesion types. Moreover, many deregulated<br />

genes <strong>of</strong> these categories could be directly associated with activated AKT, TORC1 and/or<br />

TORC2 pathways (Supplementary table 6.3 and below), and may result from a functional<br />

loss <strong>of</strong> the TSC2 gene in preneoplastic lesions (Figure 6.3, C), i.e. genes involved in: Cell<br />

cycle progression (upregulation <strong>of</strong> CDC2, CCNB1, CCNB2, AURKB; downregulation <strong>of</strong><br />

NUP50); cell growth, survival and proliferation (upregulation <strong>of</strong> CTGF, POSTN, ITGB4,<br />

PINK, IGFBP1 and PIK3AP1; downregulation <strong>of</strong> FOXOA1); protein synthesis (upregulation<br />

<strong>of</strong> RPS15A); upregulation <strong>of</strong> HIF1α target genes involved in angiogenesis (PAI1, CXCR4)<br />

and glucose uptake and glycolysis (PFKM, PFKC, ENO2, PGAM2 and GLUT2);<br />

cytoskeleton rearrangement (upregulation <strong>of</strong> genes coding for Rac/Rho/Cdc42 signal<br />

transduction proteins e.g. RHOQ, RAC2, PAK3, IQGAP1, CDC42SE1, CDC42EP1,<br />

ELMO2, DOCK6, RAC2 and PLEKHG2; upregulation <strong>of</strong> PKCα substrates MARCKS and<br />

AKAP12)<br />

Moreover, a direct correlation between functional loss <strong>of</strong> TSC2 in preneoplastic lesions and<br />

active c-myc oncogene is indicated by upregulation <strong>of</strong> C-MYC itself and its target genes<br />

EMP1 and EMP3, which may point to additional mutations up- or down-stream <strong>of</strong> the<br />

TSC2/TSC1 complex (Figure 6.3, A and C).<br />

124


Table 6.2: Numbers and categories <strong>of</strong> significantly deregulated genes in preneoplastic lesions<br />

Endpoints % <strong>of</strong> all significantly deregulated<br />

annotated genes per group<br />

bAT bAT bAT bAH bAH bAH<br />

C AA OTA C AA OTA<br />

Description<br />

Chapter 6: Manuscript IV<br />

Oxidative stress 2.9 2.7 4.7 1.9 1.4 2.4 Up regulation <strong>of</strong> ARE target genes and other genes known to be involved in oxidative stress responses. Down<br />

regulation <strong>of</strong> genes mediating the cellular antioxidant defence<br />

DNA damage<br />

response<br />

4.4 0.9 1.6 1.1 4.1 0 Up regulation <strong>of</strong> genes encoding p53 target genes and genes coding for proteins involved in DNA damage repair<br />

Apoptosis 4.3 2.4 1.6 3.4 1.4 1.2 Up regulation <strong>of</strong> pro-apoptotic genes, down regulation <strong>of</strong> anti-apoptotic genes<br />

Tumour- and<br />

metastasis<br />

suppression<br />

Cell cycle<br />

progression<br />

Cell growth/<br />

survival/<br />

proliferation<br />

2.9 3.0 4.7 2.3 2.7 3.5 Up regulation <strong>of</strong> genes coding for tumour suppressor genes or genes. Up regulation <strong>of</strong> tissue inhibitor <strong>of</strong><br />

metalloproteinases or down regulation <strong>of</strong> genes facilitating metastasis<br />

2.9 5.1 5.5 3.1 5.5 3.5 Up regulation <strong>of</strong> genes involved in DNA replication cell cycle progression, mitotic spindle/ cytokineses or<br />

nucleosome formation.<br />

10.1 8.3 8.6 6.9 6.8 10.6 Up regulation <strong>of</strong> genes encoding growth and survival factors, (IGF)-PI3K-AKT and the mTOR/S6K pathway<br />

components, anti-apoptotic genes, oncogenes. Down regulation <strong>of</strong> tumor suppressor genes.<br />

Tumourigenesis 1.4 0.9 1.6 1.1 2.7 1.2 Deregulation <strong>of</strong> genes with unclear biochemical function, however for which a similar direction <strong>of</strong> deregulation has<br />

been reported previously in different types <strong>of</strong> cancer.<br />

Cytoskeleton<br />

rearrangement<br />

Enhanced cell<br />

adhesion<br />

7.2 7.1 7.8 10.7 6.8 7.1 Up regulation <strong>of</strong> genes coding for Rho-GTPases, other proteins involved in positive regulation <strong>of</strong> actin<br />

polymerization, actin remodelling, stress fibre formation or microtubule dynamics.<br />

1.4 1.2 0.8 0.8 4.1 2.4 Up regulation <strong>of</strong> genes coding for cell adhesion molecules<br />

Angiogenesis 2.9 4.5 5.5 1.9 4.1 4.7 Up regulation <strong>of</strong> genes <strong>of</strong> the VEGF pathway and functioning in growth and survival <strong>of</strong> endothelial cells, vascular<br />

development and remodelling.<br />

125


Endpoints % <strong>of</strong> all significantly deregulated<br />

annotated genes per group<br />

bAT bAT bAT bAH bAH bAH<br />

C AA OTA C AA OTA<br />

Description<br />

Chapter 6: Manuscript IV<br />

Metastasis 1.4 2.7 2.3 3.4 2.7 1.2 Up regulation <strong>of</strong> genes coding for serin proteinases involved in ECM degradation, down regulation <strong>of</strong> genes coding<br />

for cell adhesion molecules, or gap junction proteins.<br />

Dedifferentiation 0 6.5 3.9 7.7 0 5.9 Down regulation <strong>of</strong> genes mediating the physiological homeostasis <strong>of</strong> the cell and/ or organ. Down regulation <strong>of</strong><br />

genes involved in biotransformation<br />

Stromatogenesis 2.9 4.5 6.3 7.7 5.5 12.9 Up regulation genes encoding ECM components and components involved in stroma-cell interaction like integrins or<br />

integrin binding proteins.<br />

Immune response 10.1 15.5 15.6 19.2 15.1 15.3 Up regulation <strong>of</strong> genes encoding components <strong>of</strong> the acute phase response, inflammation, innate or adaptive<br />

immunity, complement system, TNF/ cytokine pathway, or up regulation genes coding for proteins involved in<br />

antigen presentation.<br />

Lysosomal<br />

autophagy<br />

1.4 2.7 2.3 1.9 1.4 2.4 Up regulation <strong>of</strong> genes coding for lysosomal enzymes and proteins <strong>of</strong> the lysosomal membrane<br />

Energy provision 5.8 4.5 2.3 2.7 4.1 1.2 Up regulation <strong>of</strong> genes coding for proteins supporting uptake <strong>of</strong> glucose, lipids and amino acids into the cell, as well<br />

as for proteins supporting glycogenolysis, glycolysis, gluconeogenesis, citrate cycle, and glutaminolysis.<br />

Precursor provision 5.8 8.0 8.6 8.8 5.5 5.9 Up regulation <strong>of</strong> genes coding for proteins involved in fatty acid synthesis, lipid storage, cholesterol uptake,<br />

cholesterol metabolism and phospholipid / glycolipid synthesis. Down regulation <strong>of</strong> genes involved in ß-oxidation<br />

fatty acids and amino acid degradation<br />

Protein synthesis<br />

and trafficing<br />

4.3 1.2 3.1 3.4 2.7 3.5 Up regulation <strong>of</strong> genes involved in protein sorting, vesicular transport or endo-/exocytosis. Up regulation <strong>of</strong> genes<br />

involved in translation or protein folding in the endoplasmatic reticulum<br />

Protein degradation 4.3 3.3 0 0.4 0 0 Up regulation <strong>of</strong> genes coding for components <strong>of</strong> the ubiquitin-dependent protein catabolism<br />

Neuronal<br />

differentiation<br />

2.9 3.3 3.1 2.7 5.5 2.4 Up regulation <strong>of</strong> genes involved in neuronal signal transmission or neuronal differentiation<br />

Cellular stress 4.3 2.7 1.6 3.1 8.2 3.5 Up regulation <strong>of</strong> genes involved in the stress kinase-, NFkB-, or HIF pathways pathway or other stress related<br />

pathways involved in hypoxia, acidosis or tubular damage. Down regulation <strong>of</strong> genes encoding inhibitors <strong>of</strong> these<br />

pathways and endpoints<br />

Osmotic stress 7.2 1.5 2.3 1.1 1.4 1.2 Up regulation <strong>of</strong> genes involved in water and sodium resorption, down regulation <strong>of</strong> genes cell mediating volume<br />

homeostasis. Up regulation <strong>of</strong> genes involved in hyperosmotic response.<br />

Unknown endpoint 10.1 7.7 6.3 5.0 8.2 8.2 Genes with known biochemical function, but whose deregulation could not be given a specific meaning, due to the<br />

lack <strong>of</strong> information from literature<br />

126


Chapter 6: Manuscript IV<br />

6.4.5 Confirmation <strong>of</strong> TORC1 and TORC2 pathway activation in<br />

preneoplastic lesions<br />

Above presented gene expression pr<strong>of</strong>iles point to a concomitant activation <strong>of</strong> AKT,<br />

TORC1 and/or TORC2 pathways. To prove this, immunostaining <strong>of</strong> phosphorylated<br />

ribosomal protein S6 at Ser235/236 (pS6RP) was used as an indicator <strong>of</strong> TORC1 activity<br />

(Kenerson et al. 2002). Staining <strong>of</strong> AKT phosphororylated at Ser473 (pAKT) provided<br />

confirmation <strong>of</strong> active TORC2 since active TORC2 was shown to directly phosphorylate<br />

AKT at Ser473 (Sarbassov et al. 2005).<br />

Figure 6.3. Schematic overview over the postulated processes (incl. activated mTOR, cell clone<br />

survival and expansion) involved in the formation <strong>of</strong> preneoplastic lesions. (B)<br />

Postulated time curve <strong>of</strong> tissue adaptation vs. formation <strong>of</strong> preneoplastic lesions. (C)<br />

Suggested mechanistic involvement <strong>of</strong> TORC1 and TORC2 dependent mTOR<br />

pathways involved in renal carcinogenesis. Blue: pathways and processes implicated<br />

by gene expression analysis. Red: protein expression or phosphorylation verified by<br />

immunostaining.<br />

127


Chapter 6: Manuscript IV<br />

Comparable pS6RP staining was observed within bATs and bAHs <strong>of</strong> AA, OTA and control<br />

male and female Eker rats (Figure 6.4, A and B). Sporadic preneoplastic tubules were<br />

negative for pS6RP staining (a maximum <strong>of</strong> 6 bATs in the 6 months AA treatment group).<br />

Positive pS6RP staining was also present in adenomas, carcinomas and oncocytic cysts <strong>of</strong><br />

all rats examined. pS6RP positive immunostaining <strong>of</strong> preneoplastic lesions and tumors may<br />

thus be interpreted as corroborative evidence for activation <strong>of</strong> the TORC1/S6K/S6RP<br />

pathway.<br />

pAKT-immunostaining <strong>of</strong> bAT and bAH and tumors resulted in a weak but specific<br />

cytoplasmic and sporadic nuclear staining in treated and control male rats, suggesting the<br />

involvement <strong>of</strong> TORC2/AKT pathway in renal carcinogenesis (Figure 6.4, C and D).<br />

However, as pAKT staining appeared faint, additional immunostaining <strong>of</strong> AKT target<br />

FOXO1 was employed, since FOXO1 shuttles from the nucleus to cytoplasm for<br />

degradation when phosphorylated by active AKT (Tran et al. 2003). Since, FOXO1<br />

phosphorylation was demonstrated to be strongly reduced in mice lacking TORC2 member<br />

rictor (Jacinto et al. 2006), cytoplasmatic localization <strong>of</strong> FOXO1 can be interpreted as<br />

TORC2 dependent AKT activation. Healthy tubules presented primarily with nuclear<br />

FOXO1 staining. In contrast, strong cytoplasmic and nuclear staining in bATs and bAHs<br />

was detected, as expected for deregulated FOXO1 activation and/or half-life (Figure 6.4, E<br />

and F). Thus, the FOXO1 staining pattern further supports activation <strong>of</strong> the TORC2-AKT<br />

pathway in preneoplastic renal lesions.<br />

128


Chapter 6: Manuscript IV<br />

Figure 6.4: Pro<strong>of</strong> <strong>of</strong> principle staining for concurrent TORC1 and TORC2 activation in<br />

preneoplastic lesions. Serial sectioned bAT with positive pS6RP (A), pAKT (C) and<br />

cytoplasmatic accumulation <strong>of</strong> FOXO1 (E). Representative images <strong>of</strong> bAH<br />

immunopositive for pS6RP (B), p-AKT (D), and FOXO1 (F). Additional images <strong>of</strong><br />

pS6RP, p-AKT and FOXO1 are shown in Figure S3<br />

129


6.5 Discussion<br />

Chapter 6: Manuscript IV<br />

6.5.1 Compound-induced tubular damage and regenerative cell<br />

proliferation is critical for the formation but not for the progression <strong>of</strong><br />

preneoplastic lesions<br />

Increased cell proliferation can result in carcinogenesis, most likely by the fixation <strong>of</strong><br />

mutations as well as by survival and promotion <strong>of</strong> initiated cells (Barrett and Wiseman<br />

1987; Dietrich and Swenberg 1991). By distinguishing nephron specific effects and<br />

“healthy” from pathologically altered nephron-sections, above data demonstrated that AAand<br />

OTA-induced increase <strong>of</strong> cell proliferation is restricted to pathologically altered<br />

proximal tubules as well as to preneoplastic lesions at the time points evaluated.<br />

If compound induced nephrotoxicity/genotoxicity and accompanying regenerative cell<br />

proliferation is a main contributor to renal carcinogenesis, compound-induced cell<br />

proliferation should correspond to increased numbers and/or progression rates <strong>of</strong><br />

preneoplastic lesions in a compound- and sex-specific manner. Indeed the 2-fold higher<br />

cell proliferation in OTA-treated males than in females at the 6-month time-point<br />

corresponded well with the 1.7-fold higher increase <strong>of</strong> preneoplastic lesions in males than<br />

in females (Figure 6.1, Supplementary table 6.2). Since ratios <strong>of</strong> bAT and bAH numbers<br />

remained unchanged, the 80% incidence <strong>of</strong> bAHs in males compared to the 30% in<br />

females (Figure 6.1, D) suggest an earlier onset <strong>of</strong> preneoplastic lesion development in<br />

males and thus points to a sex-dependent difference in OTA-induced formation <strong>of</strong><br />

preneoplastic lesions rather than to an OTA-mediated lesion progression. The sexdependent<br />

difference in formation <strong>of</strong> preneoplastic lesions, as reported here, may result<br />

from a higher OTA-induced oxidative stress and subsequent DNA damage in male rats<br />

(Kamp et al. 2005). This interpretation is supported by the finding that 14-days oral<br />

treatment <strong>of</strong> Eker and wild type female rats with OTA resulted in an upregulation <strong>of</strong> the 8oxoguanine<br />

DNA glycosylase (an enzyme critically involved in the repair <strong>of</strong> oxidative DNA<br />

damage) than in the corresponding males (Supplementary figure 6.4). Moreover, catalase<br />

expression was reduced in OTA-treated male Eker and wild type rats, while females<br />

showed an increased expression.<br />

The critical role <strong>of</strong> compound induced nephrotoxicity/genotoxicity and accompanying cell<br />

proliferation for the onset <strong>of</strong> preneoplastic lesion formation is corroborated by the findings in<br />

the AA-treatment groups. Indeed, the 2-fold higher cell proliferation in female than in male<br />

rats after 3 months <strong>of</strong> AA-treatment corresponded to the 2-fold higher number <strong>of</strong><br />

preneoplastic lesions in females at the same time point. Moreover, a similar sex-dependent<br />

130


Chapter 6: Manuscript IV<br />

trend for cell proliferation and preneoplastic lesions, although not as pronounced, was also<br />

observed at the 6 months time-point (Figure 6.1, Supplemetary figure 6.2, Table 6.1 and<br />

Supplemetary table 6.2). As noted for OTA-treated rats, the comparable bAT to bAH ratio in<br />

male and female AA-treated rats is suggestive <strong>of</strong> only a limited influence <strong>of</strong> AA on bAT to<br />

bAH progression. The sex-dependent difference in the formation <strong>of</strong> AA-induced lesions<br />

may result from the previously reported 2-fold higher levels <strong>of</strong> dG-AAI and dA-AAII DNA<br />

adducts in kidneys <strong>of</strong> female than male Wistar rats following treatment with oral doses <strong>of</strong><br />

0.15mg/kg body weight for 3 months (Arlt et al. 2001). Given a comparable cellular uptake<br />

and metabolism <strong>of</strong> AA in Eker and Wistar rats, the higher number <strong>of</strong> DNA adducts and<br />

regenerative cell proliferation observed in female rats may have led to a higher fixation <strong>of</strong><br />

mutations and therefore explain the observed corresponding formation <strong>of</strong> preneoplastic<br />

lesions.<br />

6.5.2 Time dependent tissue adaptation to carcinogen treatment<br />

Gene expression analysis <strong>of</strong> preneoplastic lesions demonstrated that bATs and bAHs <strong>of</strong><br />

control, AA- and OTA-treated male rats were qualitatively indistinguishable (Figure 6.2),<br />

thus supporting the above assumption that once initiated, in a compound- and sexdependent<br />

manner, clonal expansion <strong>of</strong> preneoplastic lesions is compound-independent.<br />

Moreover, while 14-days AA- and OTA- treatment <strong>of</strong> Eker and corresponding wild type rats<br />

resulted in substantial gene expression changes in the renal cortex, e.g. response to<br />

oxidative stress and DNA damage (Stemmer et al. 2007), only marginal gene expression<br />

changes were observed in healthy tubules <strong>of</strong> male rats following 6 months AA- and OTAtreatment<br />

(Figure 6.2). This suggests that part <strong>of</strong> the renal cells may have the potential to<br />

adapt to compound treatment, thus further supporting the observation that compounds are<br />

important for the formation but not the clonal expansion <strong>of</strong> preneoplastic lesions. These<br />

findings thus imply the existence <strong>of</strong> a critical, albeit compound-specific, period <strong>of</strong> exposure<br />

for genotoxic, or non-genotoxic carcinogens (Figure 6.3, B), where normal renal tissue is<br />

not yet adapted. In this critical period normal renal tissue appears incapable <strong>of</strong> sufficiently<br />

repairing compound-induced damage despite active regenerative cell proliferation, thus<br />

providing the possibility for fixation <strong>of</strong> mutations and thereby formation <strong>of</strong> preneoplastic<br />

lesions.<br />

131


Chapter 6: Manuscript IV<br />

6.5.3 Numbers <strong>of</strong> earliest preneoplastic lesions formed are decisive for<br />

later occurrence <strong>of</strong> veritable tumors<br />

The existence <strong>of</strong> a critical period <strong>of</strong> carcinogen exposure would imply, that extended<br />

compound exposure may add little to the formation <strong>of</strong> additional preneoplastic lesions<br />

(Figure 6.3, B), thus suggesting that the incidence and number <strong>of</strong> veritable tumors is<br />

primarily driven by the number <strong>of</strong> preneoplastic lesions formed during this critical period.<br />

Indeed, the incidences <strong>of</strong> preneoplastic lesions (bAH) observed in the Eker rat study (80%<br />

in males vs. 30% in females) correspond well to the tumor incidences observed at the end<br />

<strong>of</strong> the OTA 2-year bioassay (87.2% in male versus 22.9% in female F344 rats) at the same<br />

dose (Boorman et al. 1992). This could imply that the incidence and number <strong>of</strong> veritable<br />

tumors is primarily driven by the number <strong>of</strong> preneoplastic lesions initially formed during a<br />

critical period <strong>of</strong> carcinogen exposure (see above). This interpretation is supported by study<br />

<strong>of</strong> Wolf and co-workers who reported an indistinguishable number <strong>of</strong> preneoplastic lesions<br />

and tumors in Eker rats treated with 500 ppm sodium barbital (NaBB) in feed for 4.5<br />

months and sacrificed at 12 months and Eker rats treated with 500 ppm NaBB in feed for<br />

the whole 12 months (Wolf et al. 2000). The latter phenomenon is not restricted to Eker<br />

rats, as confirmed by a previous study (Dietrich and Swenberg 1991), where male NBR<br />

rats, deficient for α2u-globulin and male F344 were initiated with the genotoxic N-ethyl-N-<br />

hydroxyethylnitrosamine (EHEN) and/or promoted with α2u-globulin binding d-limonene for<br />

6 months. D-limonene exposure was demonstrated to induce concentration-dependent<br />

regenerative cell proliferation in F344 but not in the α2u-globulin deficient NBR rats. Both<br />

EHEN and d-limonene treatment resulted in the formation <strong>of</strong> preneoplastic lesions (AT and<br />

AH) in F344 rats in nearly comparable numbers but not in the formation <strong>of</strong> veritable tumors.<br />

Similarly, treatment <strong>of</strong> NBR rats with EHEN or EHEN and d-limonene led to comparable<br />

numbers <strong>of</strong> ATs and AHs but no tumors. In contrast, treatment <strong>of</strong> F344 rats with a<br />

combination <strong>of</strong> EHEN and d-limonene led to highest number <strong>of</strong> ATs and AHs as well as to<br />

the formation <strong>of</strong> veritable tumors.<br />

The latter data thus strongly support the observation that the number <strong>of</strong> preneoplastic<br />

lesions initially formed, resulting from genotoxicity, fixation <strong>of</strong> spontaneous mutations via a<br />

mitogenic stimulus or regenerative cell proliferation or a combination there<strong>of</strong> (Figure 6.3, A<br />

and B), are decisive for the later development <strong>of</strong> renal tumors in rats.<br />

132


Chapter 6: Manuscript IV<br />

6.5.4 The balance <strong>of</strong> TORC1 and TORC2 pathways determines<br />

preneoplastic lesion progression<br />

Since carcinogen treatment may mainly affect early tumor formation in a compound- and<br />

sex-specific manner, similar gene expression pr<strong>of</strong>iles in preneoplastic lesions <strong>of</strong> AA, OTA<br />

and control Eker rats may point to a central pathway involved in lesion progression.<br />

Functional analysis <strong>of</strong> deregulated genes in bATs and bAHS as well as<br />

immunohistochemical staining <strong>of</strong> pS6RP, pAKT and FOXO1 in bATs and bAHs <strong>of</strong> control,<br />

AA- and OTA-treated Eker rats clearly demonstrated that both mTOR pathways (TORC1<br />

and TORC2 dependant) were activated. The latter may result from a functional loss <strong>of</strong> the<br />

TSC2 gene in preneoplastic lesions either via loss-<strong>of</strong> heterozygosity (Yeung et al. 1995) or<br />

additional mutations up- or down-stream <strong>of</strong> the TSC2/TSC1 complex e.g. the c-myc<br />

oncogene (Figure 6.3, A and C). Eker rat preneoplastic lesion analysis demonstrated an<br />

upregulation <strong>of</strong> several HIF1α �target genes (Supplemetary table 6.3) and numerous<br />

additional genes involved in downstream responses <strong>of</strong> HIF1α e.g. angiogenesis and<br />

energy provision (Table 6.2 and S3). The latter findings correspond well to the role <strong>of</strong><br />

activated TORC1 shown to regulate several cell growth regulators, including stabilization <strong>of</strong><br />

the transcription factor HIF1α (Brugarolas et al. 2003), thereby providing for sufficient<br />

oxygen and energy supply <strong>of</strong> proliferating cells, e.g. via angiogenesis and glycolysis<br />

(Semenza 2002). Under physiological conditions, activated S6K leads to phosphorylation<br />

and degradation <strong>of</strong> insulin receptor substrate proteins (Harrington et al. 2004; Shah et al.<br />

2004) and consequently to reduced downstream PI3K-AKT-TSC1/2 activation in<br />

mammalian cells. Since intact S6K dependent negative feedback inhibition and limited<br />

oncogenic PI3K-AKT signaling was still present in growth-limited benign hamartomas<br />

(Kwiatkowski 2003; Manning et al. 2005), continued and uncontrolled activation <strong>of</strong> both<br />

S6K and AKT could be a critical trigger in the progression <strong>of</strong> benign to rapidly growing<br />

malignant lesions.<br />

The concomitant activation <strong>of</strong> TORC1 and TORC2 (Figure 6.3, C and 6.4) and therefore<br />

likely disturbance <strong>of</strong> feedback inhibition <strong>of</strong> mTOR-AKT signaling in renal preneoplastic<br />

lesions, as shown here, appears important for survival and clonal expansion <strong>of</strong><br />

preneoplastic lesions. Facilitated AKT activation due to TORC2 dependent Ser473<br />

phosphorylation (Bellacosa et al. 2005), would not only further enhance the AKT-TSC2-<br />

TORC1 pathway, but also influence other targets <strong>of</strong> the oncogenic AKT leading to reduced<br />

apoptosis and cell cycle progression (Table 6.2 and S3).<br />

Moreover, preneoplastic lesion gene expression pr<strong>of</strong>iles support the previously shown<br />

function <strong>of</strong> TORC2 as an important regulator <strong>of</strong> the cytoskeleton through members <strong>of</strong> the<br />

133


Chapter 6: Manuscript IV<br />

Rho small GTPase family and phosphorylation <strong>of</strong> Protein Kinase Cα (PKCα) (Jacinto et al.<br />

2004; Sarbassov et al. 2004), which were discussed to play a role in cell proliferation,<br />

transformation and metastasis (Jaffe and Hall 2002). TORC2 dependent AKT<br />

phosphorylation and upregulation <strong>of</strong> members <strong>of</strong> the Rho small GTPase family however<br />

also suggest that TORC2 may be important initial formation <strong>of</strong> preneoplastic lesions.<br />

Indeed, Kenerson and co-workers (Kenerson et al. 2005), demonstrated that rapamycin<br />

inhibited renal preneoplastic progression to veritable renal tumors in Eker rats but not the<br />

formation <strong>of</strong> earliest preneoplastic lesions, suggesting that the balance between rapamycin<br />

sensitive TORC1 and the insensitive TORC2 may be critical for preneoplastic lesion<br />

survival and clonal expansion. Rapamycin treatment may inhibit only the TORC1/HIF1α<br />

dependent precursor-, energy- and oxygen supply, necessary for rapid clonal expansion <strong>of</strong><br />

larger preneoplastic renal lesions to veritable tumors, while TORC2/AKT dependent cell<br />

cycle progression and apoptosis inhibition may be important for the survival and<br />

proliferation <strong>of</strong> initiated cells and earliest preneoplastic lesions.<br />

6.5.5 Outlook<br />

In view <strong>of</strong> the current interest in the use <strong>of</strong> rapamycin, and analogous inhibitors <strong>of</strong> the<br />

mTOR pathway for treatment <strong>of</strong> human renal cancer and the close analogy <strong>of</strong> rat and<br />

human renal tumors, the results presented here suggest that TORC1 inhibitors (e.g.<br />

rapamycin analogues) would reduce the rate <strong>of</strong> preneoplastic to neoplastic progression but<br />

not the origin <strong>of</strong> the disease. Concurrent treatment with rapamycin analogues and a<br />

specific TORC2 inhibitor thus holds the promise that growth and survival <strong>of</strong> both the initial<br />

preneoplastic lesions and advanced progression stages could be specifically affected.<br />

Acknowledgements<br />

We would like to thank the Federal Ministry <strong>of</strong> Education and Research for funding the<br />

project (BMBF: 0313024), T. Lampertsdörfer, G. von Scheven, A. Heussner and E. O’Brien<br />

for help with the animal experiment, M. Thiel and K. Lotz for microarray hybridization and P.<br />

Pfluger for critically reading the manuscript.<br />

134


6.6 Supplemental Material<br />

6.6.1 Supplementary experimental procedures: Real time PCR<br />

Chapter 6: Manuscript IV<br />

To evaluate gene expression by real time PCR, total RNA was extracted from frozen renal<br />

cortex <strong>of</strong> 14-days OTA treated and control male and female Eker and wild type rats (n=3)<br />

using RNEasy Mini Kit (Qiagen), according to manufacturer’s instructions. Following DNase<br />

treatment, reverse transcriptions were performed using SuperScript III (Invitrogen) and<br />

random primers (Invitrogen). Real-time PCRs <strong>of</strong> the house keeping gene 18S RNA and <strong>of</strong><br />

candidate genes 8-oxoguanine DNA glycosylase (OGG1) and catalase (CAT) were<br />

performed on Biorad iCycler using iQ SybrGreen Supermix (Biorad). All samples were<br />

performed in technical duplicates. Relative amounts <strong>of</strong> gene-specific mRNAs were<br />

calculated via Ct values, based on a standard curve <strong>of</strong> five points with known amounts <strong>of</strong><br />

template cDNA.<br />

135


6.6.2 Supplementary Figures<br />

Chapter 6: Manuscript IV<br />

Supplementary figure 6.1: Overview over the experimental design: The experiments were<br />

performed according to the German Animal Protection Law, approved by the relevant<br />

German authority, the Regierungspräsidium in Freiburg, Germany (registry number:<br />

G-03)<br />

136


Chapter 6: Manuscript IV<br />

Supplementary figure 6.2: H&E stained paraffin sections: (A) proximal tubules with a normal (nonpathologic)<br />

phenotype (healthy tubules; HT); (B) chronic progressive nephropathy;<br />

(C-F) representative BrdU staining patterns <strong>of</strong> different tubular sections: Red nuclei:<br />

BrdU positive, blue nuclei: haematoxylin counterstaining; (C) proximal tubules <strong>of</strong> the<br />

outer renal cortex (P1/P2 segments); (D) proximal tubules <strong>of</strong> the inner renal cortex (P3<br />

segments); (E) distal tubules; and (F) collecting duct.<br />

137


Chapter 6: Manuscript IV<br />

Supplementary figure 6.3; Representative immunmohistochemical staining <strong>of</strong> renal paraffin<br />

sections demonstrating positive pS6RP (A), pAKT (B) and cytoplasm accumulation <strong>of</strong><br />

Foxo1 (C) demonstrate the concurrent activation <strong>of</strong> both mTOR pathways.<br />

Representative images <strong>of</strong> bAHs positively with antibodies against pS6RP (D), pAKT<br />

(E) and Foxo1 (F).<br />

138


Chapter 6: Manuscript IV<br />

Supplementary figure 6.4: Gene expression analysis <strong>of</strong> OGG1 and CAT in male and female Eker<br />

and wild type rats following 14-days treatment with ochratoxin A (Stemmer et al.<br />

2007). Data <strong>of</strong> three replicate animals are shown as mean + SD. Newman-Keuls<br />

Multiple Comparison Test was performed to test for significance: * < 0.05, ** < 0.01.<br />

139


Supplemental Tables<br />

Chapter 6: Manuscript IV<br />

Supplementary table 6.1: Non-neoplastic renal pathology <strong>of</strong> Eker rats treated with AA or OTA for<br />

three and six months respectively. Histopathological changes were ranked from none<br />

(0) to severe (4) including intermediate classes. Values are presented as median ±<br />

median absolute deviation.<br />

Male Eker rats 3 months 6 months<br />

Control AA OTA Control AA OTA<br />

Necrosis 0.0 + 0.0 0.0 + 0.1 0.0 + 0.2 0.0 + 0.0 0.0 + 0.2 0.8 + 0.6 b<br />

Apoptosis 0.0 + 0.0 0.0 + 0.1 1.0 + 0.2 a 0.0 + 0.0 0.0 + 0.2 0.3 + 0.4 a<br />

Karyomegaly 0.0 + 0.0 0.0 + 0.2 2.0 + 0.5 a 0.0 + 0.0 0.0 + 0.1 1.8 + 0.5 c<br />

Vacuolization 0.0 + 0.0 0.0 + 1.0 0.0 + 0.4 0.0 + 0.0 0.3 + 0.7 1.5 + 0.9 b<br />

Cell shedding 1.0 + 0.6 1.0 + 0.3 1.3 + 0.5 1.0 + 0.6 1.0 + 0.6 0.5 + 0.4<br />

Protein casts 0.3 + 0.6 0.5 + 0.4 1.0 + 0.6 1.0 + 0.5 1.8 + 0.7 1.3 + 0.9<br />

Tubular dilatation 0.5 + 0.5 0.5 + 0.3 1.0 + 0.6 0.5 + 1.0 1.0 + 0.4 1.0 + 0.8<br />

Calcium Casts 0.0 + 0.4 0.0 + 0.0 0.0 + 0.0 0.0 + 0.0 0.0 + 0.0 0.0 + 0.0<br />

Regeneration 0.5 + 0.2 0.5 + 0.3 1.0 + 0.3 0.0 + 0.5 1.0 + 0.6 1.0 + 0.5 a<br />

CPN 0.5 + 0.2 0.5 + 0.1 1.0 + 0.3 a 0.8 + 0.5 1.5 + 0.4 a 2.0 + 0.3 b<br />

Inflammation 0.3 + 0.3 0.5 + 0.2 0.5 + 0.3 0.5 + 0.4 1.0 + 0.6 a 1.3 + 0.6 a<br />

Deposit <strong>of</strong> pigments 0.1 + 0.2 0.0 + 0.1 0.0 + 0.3 0.0 + 0.1 0.0 + 0.2 0.3 + 0.4<br />

Female Eker rats 3 months 6 months<br />

Control AA OTA Control AA OTA<br />

Necrosis 0.0 + 0.0 0.0 + 0.1 0.0 + 0.1 0.0 + 0.3 1.0 + 0.6 1.3 + 0.6 b<br />

Apoptosis 0.0 + 0.0 0.0 + 0.1 0.5 + 0.2 c 0.0 + 0.0 0.0 + 0.2 0.0 + 0.2<br />

Karyomegaly 0.0 + 0.1 0.0 + 0.2 1.0 + 0.8 c 0.0 + 0.0 0.0 + 0.3 1.3 + 0.6 c<br />

Vacuolization 0.0 + 0.2 0.0 + 0.0 0.0 + 0.0 2.0 + 0.4 2.3 + 0.9 2.5 + 0.4 b<br />

Cell shedding 1.0 + 0.3 1.0 + 0.4 1.3 + 0.6 1.0 + 0.4 1.0 + 0.5 1.0 + 0.5<br />

Protein casts 0.5 + 0.5 0.5 + 0.3 0.8 + 0.5 0.0 + 0.7 1.8 + 0.9 1.5 + 0.6<br />

Tubular dilatation 1.0 + 0.6 0.3 + 0.7 1.0 + 0.6 0.5 + 0.5 1.0 + 1.0 1.0 + 0.5<br />

Calcium Casts 0.0 + 0.2 0.0 + 0.4 0.0 + 0.1 0.0 + 0.0 0.0 + 0.0 0.0 + 0.1<br />

Regeneration 0.3 + 0.4 0.0 + 0.3 0.0 + 0.4 0.5 + 0.5 1.0 + 0.6 1.0 + 0.5<br />

CPN 0.5 + 0.2 0.5 + 0.3 1.5 + 0.3 c 1.0 + 0.3 1.3 + 0.5 1.5 + 0.3 a<br />

Inflammation 0.5 + 0.2 0.5 + 0.2 0.5 + 0.4 0.5 + 0.3 0.5 + 0.2 1.0 + 0.2 a<br />

Deposit <strong>of</strong> pigments 0.1 + 0.2 0.0 + 0.1 0.0 + 0.2 0.0 + 0.3 0.3 + 0.4 0.0 + 0.2<br />

a b<br />

Significantly higher than the respective control group. Mann-Whitney test (p< 0.05). Significantly higher<br />

than the respective control group. Mann-Whitney test (p< 0.005). c Significantly higher than the respective<br />

control group. Mann-Whitney test (p< 0.001)<br />

140


Chapter 6: Manuscript IV<br />

Supplementary table 6.2: (Pre-) neoplastic pathology <strong>of</strong> the renal cortex <strong>of</strong> male and female Eker<br />

rats treated with AA or OTA for three and six months respectively. Incidence and<br />

numbers <strong>of</strong> bAT, bAH and microscopic (m) and gross (g) tumours (adenoma and<br />

carcinoma) are displayed. Ratios <strong>of</strong> bATs and bAHs and tumours per rat are<br />

presented as mean + standard deviation. Additional ratios <strong>of</strong> mean bAT/rat to mean<br />

bAH/rat are indicative for a bAT to bAH progression.<br />

bAT<br />

bAH<br />

Tumor<br />

bAT<br />

bAH<br />

Tumor<br />

Male Eker rats 3 months 6 months<br />

Control AA OTA Control AA OTA<br />

Incidence 10/10 10/10 10/10 10/10 10/10 10/10<br />

Outer cortex 15 27 32 20 66 42<br />

Cortex/Medulla 20 28 84 30 107 107<br />

Total bAT 35 55 116 50 173 150<br />

bAT / rat 3.5 + 2.0 5.5 + 2.6 11.6+2.0 a 5.0 + 3.3 17.3+5.7 a 15.0+2.2 a<br />

Incidence 4/10 5/10 5/10 5/10 8/10 8/10<br />

Outer cortex 1 1 3 1 16 9<br />

Cortex/Medulla 4 4 6 10 15 17<br />

Total 5 5 9 11 31 26<br />

bAH / rat 0.6 + 0.9 0.5 + 0.5 0.9 + 1.0 1.1 + 1.5 3.1 + 2.8 2.6 + 1.9<br />

Ratio 5.8 9.2 12.9 4.5 5.5 5.7<br />

Incidence (m) 0/10 3/10 1/10 1/10 3/10 2/10<br />

Total (m) 0 3 1 1 3 2<br />

Total (g + m) 1 3 1 1 3 5<br />

Tumour / rat 0.0 + 0.0 0.3 + 0.7 0.1 + 0.3 0.1 + 0.3 0.3 + 0.5 0.2 + 0.4<br />

Female Eker rats 3 months 6 months<br />

Incidence 10/10 10/10 10/10 9/9 8/8 10/10<br />

Outer cortex 16 46 24 16 67 36<br />

Cortex/Medulla 19 61 76 12 63 54<br />

Total bAT 35 107 100 28 130 90<br />

bAT / rat 3.5 + 1.7 10.7+6.2 a 10.0+3.3 a 3.1 + 1.3 16.3+4.8 a 9.0 + 3.8 a<br />

Incidence 1/10 3/10 5/10 2/9 7/8 c 3/10<br />

Outer cortex 0 4 0 2 13 2<br />

Cortex/Medulla 1 2 5 0 6 5<br />

Total 1 6 5 2 19 7<br />

bAH / rat 0.1 + 0.3 0.6 + 1.0 0.5 + 0.5 0.2 + 0.4 2.4 + 1.6 b 0.7 + 1.5<br />

Ratio 35 17.8 20.0 15.5 6.8 12.9<br />

Incidence (m) 3/10 0/10 0/10 1/10 1/10 3/10<br />

Total (m) 3 0 0 1 1 3<br />

Total (g + m) 3 0 0 1 2 4<br />

Tumour / rat 0.3 + 0.5 0.0 + 0.0 0.0 + 0.0 0.1 + 0.3 0.1 + 0.6 0.3 + 0.5<br />

a b<br />

Significantly higher than the respective control group. Dunnett`s test (p< 0.01). Significantly higher than<br />

the respective control group. Dunnett`s test (p< 0.05). c Significantly higher incidence than the respective<br />

control group. Fisher`s exact test (p< 0.05). d Mortality: One female control animal died 2 weeks before<br />

sacrifice. Two female AA treated Eker rats died 10 and 3 weeks before sacrifice.<br />

141


Chapter 6: Manuscript IV<br />

Supplementary table 6.3: Genes differentially deregulated in preneoplastic lesions compared to HT <strong>of</strong> control animals. For the major signalling pathways<br />

(activated (+) or inhibited (-)), genes are listed together with their Genebank accession number and the suspected pathophysiological<br />

consequence <strong>of</strong> deregulation (endpoint). The mean fold deregulation + SD are given for every lesion type. Significant deregulations vs. HT(C)<br />

were tested by Student’s t- test: b p< 0.005; c p< 0.001 which matched the p-value cut-<strong>of</strong>f <strong>of</strong> 0.005 and 2.0 fold deregulation cut-<strong>of</strong>f. (a)<br />

Significantly deregulated (p< 0.05), however not matching the chosen p-value cut<strong>of</strong>f


Accession<br />

number<br />

Protein synthesis NM_053982 RPS15A Ribosomal protein<br />

S15a<br />

Angiogenesis<br />

Glucose uptake<br />

and glycolysis<br />

Cytoskeleton<br />

rearrangement<br />

NM_012620 PAI1 Plasminogen activator<br />

inhibitor-1<br />

AA945737 CXCR4 CXC chemokine<br />

receptor 4<br />

NM_031715 PFKM 6-phosph<strong>of</strong>ructokinase,<br />

muscle (PFK-A)<br />

BM389769 PFKC 6-phosph<strong>of</strong>ructokinase,<br />

type C (PFKP)<br />

AF019973 ENO2 Enolase 2 (Gamma<br />

enolase)<br />

NM_017328 PGAM2 Phosphoglycerate<br />

mutase 2<br />

NM_012879 GLUT2 Glucose transporter<br />

type 2, liver (SLC2A2)<br />

NM_022590 SGLT2 Sodium/glucose<br />

cotransporter2<br />

AA849961 RHOQ Ras homolog gene<br />

family, member Q<br />

NM_019210 PAK3 p21-activated kinase 3<br />

(PAK beta)<br />

BM387072 IQGAP1 IQ motif containing<br />

GTPase activating protein 1<br />

BI275583 CDC42EP1 CDC42 effector<br />

protein<br />

BM389459 CDC42SE1 CDC42 small<br />

effector 2<br />

BM386781 ELMO2 Engulfment and cell<br />

motility protein 2<br />

AI102881 DOCK6 Dedicator <strong>of</strong> cytokinesis<br />

6 (predicted)<br />

Gene title References (Pub Med<br />

ID)<br />

PMID: 18092230<br />

PMID: 16990457<br />

Chapter 6: Manuscript IV<br />

Fold deregulation vs. HT control (mean and p-value <strong>of</strong> 3 replicate animals per<br />

lesion)<br />

1.04+<br />

0.15<br />

PMID: 18381294 1.41+<br />

0.96<br />

PMID: 18375114<br />

PMID: 15802268<br />

1.06+<br />

0.32<br />

PMID: 17661163 1.20+<br />

0.28<br />

PMID: 17661163 1.50+<br />

0.27<br />

PMID: 16227210 1.62+<br />

0.62<br />

PMID: 9688259 0.79+<br />

0.32<br />

PMID: 17203215 2.13+<br />

0.30 (a)<br />

PMID: 17581928 2.58+<br />

0.67<br />

PMID: 12234610<br />

PMID: 15467718<br />

1.03+<br />

0.26<br />

PMID: 18054038 0.65+<br />

0.35<br />

PMID: 15890984<br />

PMID: 15890984<br />

1.60+<br />

0.36<br />

NETAFFX 1.11+<br />

0.13<br />

PMID: 10816584 1.26+<br />

0.04 (a)<br />

PMID: 12879077 1.11+<br />

0.25<br />

PMID: 18291711<br />

PMID: 17196961<br />

1.67+<br />

0.03<br />

1.22+<br />

0.11<br />

4.19+<br />

3.74<br />

0.66+<br />

0.37<br />

1.13+<br />

0.67<br />

1.49+<br />

0.67<br />

1.72+<br />

0.65<br />

5.03+<br />

3.47<br />

2.43+<br />

0.49 (a)<br />

1.52+<br />

0.36<br />

1.07+<br />

0.10<br />

0.86+<br />

0.76<br />

1.11+<br />

0.50<br />

0.71+<br />

0.12<br />

1.06+<br />

0.28<br />

1.12+<br />

0.10<br />

1.48+<br />

0.18 (a)<br />

1.48+<br />

0.45<br />

5.85+<br />

2.45 (a)<br />

2.06+<br />

0.31 b<br />

2.54+<br />

0.33 b<br />

3.30+<br />

0.60 (a)<br />

1.79+<br />

0.61<br />

24.40+<br />

5.76 (a)<br />

2.99+<br />

1.39<br />

3.21+<br />

1.71<br />

1.31+<br />

0.15 (a)<br />

0.91+<br />

0.09<br />

3.89+<br />

0.80 b<br />

2.28+<br />

0.70 a<br />

2.24+<br />

0.39 b<br />

2.01+<br />

0.55 (a)<br />

1.44+<br />

0.22<br />

1.95+<br />

0.15 b<br />

4.70+<br />

2.74<br />

1.79+<br />

0.44 (a)<br />

1.60+<br />

0.16 (a)<br />

4.51+<br />

0.56 b<br />

2.22+<br />

0.30 b<br />

6.02+<br />

2.90<br />

11.25+<br />

1.34 c<br />

4.29+<br />

0.77 b<br />

2.36+<br />

0.24 c<br />

14.85+<br />

3.14 c<br />

3.24+<br />

0.7 (a)<br />

1.89+<br />

0.27 (a)<br />

2.45+<br />

0.22 c<br />

2.01+<br />

0.09 b<br />

1.55+<br />

0.25<br />

1.81+<br />

0.44 (a)<br />

13.72+<br />

1.70 c<br />

1.68+<br />

0.25<br />

0.93+<br />

0.22<br />

2.40+<br />

0.25<br />

2.83+<br />

0.88 (a)<br />

1.68+<br />

1.16<br />

13.43+<br />

3.14 c<br />

2.48+<br />

0.10<br />

1.67+<br />

0.33 (a)<br />

6.20+<br />

2.22 (a)<br />

1.75+<br />

0.20<br />

0.84+<br />

0.08<br />

1.77+<br />

0.13 (a)<br />

1.59 +<br />

0.14 (a)<br />

1.73+<br />

0.14 (a)<br />

1.85+<br />

0.48<br />

7.79+<br />

3.17 b<br />

3.11+<br />

0.71 b<br />

1.68+<br />

0.35 (a)<br />

3.09+<br />

0.71 (a)<br />

2.75+<br />

0.43 b<br />

9.54+<br />

2.61 (a)<br />

6.58+<br />

1.16 c<br />

3.51+<br />

1.94 a<br />

1.82+<br />

0.80<br />

8.28+<br />

9.93<br />

3.32+<br />

0.31 b<br />

2.04+<br />

0.27 b<br />

2.21 +<br />

0.22 c<br />

2.23+<br />

0.19 b<br />

1.68+<br />

0.09<br />

1.76+<br />

0.33 (a)<br />

5.34+<br />

3.53<br />

1.41+<br />

0.20 (a)<br />

1.65+<br />

0.32<br />

3.48+<br />

0.69 (a)<br />

2.74+<br />

0.44 b<br />

5.54+<br />

3.54<br />

6.84+<br />

0.84 c<br />

2.25+<br />

0.07 a<br />

1.72+<br />

0.35 (a)<br />

9.82+<br />

3.36 b<br />

2.76+<br />

0.10 b<br />

1.54+<br />

0.18 (a)<br />

2.11+<br />

0.39 (a)<br />

1.65 +<br />

0.28 (a)<br />

1.46+<br />

0.15<br />

2.29+<br />

0.28 b<br />

16.96+<br />

8.25 b<br />

1.97+<br />

0.47<br />

0.85+<br />

0.13<br />

2.22+<br />

0.26<br />

2.83+<br />

0.47 (a)<br />

0.30+<br />

0.12<br />

11.28+<br />

6.21 b<br />

1.97+<br />

0.26<br />

2.01+<br />

0.45 (a)<br />

4.76+<br />

3.36<br />

1.76+<br />

0.12<br />

0.94+<br />

0.16<br />

1.62+<br />

0.29<br />

1.73 +<br />

0.39 (a)<br />

2.11+<br />

0.11 b<br />

143


C-MYC<br />

Accession<br />

number<br />

AI010476 RAC2 Ras-related C3 botulinum<br />

toxin substrate 2 (p21-Rac2)<br />

BF397719 PLEKHG2 Pleckstrin homology<br />

domain containing G2<br />

NM_030862 MARCKS Myristoylated alaninerich<br />

protein kinase C substrate<br />

BG663107 AKAP12 A kinase anchor<br />

protein 12 (gravin)<br />

NM_012603 CMYC Myc proto-oncogene (cmyc)<br />

BI275741 EMP1 Epithelial membrane<br />

protein 1 (TMP Tumorassociated<br />

membrane protein)<br />

NM_030847 EMP3 Epithelial membrane<br />

protein 3<br />

Gene title References (Pub Med<br />

ID)<br />

PMID: 18230340<br />

PMID: 10843388<br />

Chapter 6: Manuscript IV<br />

Fold deregulation vs. HT control (mean and p-value <strong>of</strong> 3 replicate animals per<br />

lesion)<br />

1.34+<br />

0.39<br />

PMID: 18045877 1.52+<br />

0.21 (a)<br />

PMID: 16109477 1.13+<br />

0.13<br />

PMID: 11282019 1.09+<br />

0.40<br />

PMID: 18056446<br />

PMID: 12894220<br />

PMID: 18056446<br />

PMID: 12894220<br />

PMID: 18056446<br />

PMID: 12894220<br />

1.50+<br />

0.23<br />

1.18+<br />

0.19<br />

1.21+<br />

0.40<br />

1.26+<br />

0.36<br />

1.36+<br />

0.12<br />

0.83+<br />

0.29<br />

2.50+<br />

2.16<br />

1.07+<br />

0.36<br />

1.22+<br />

0.53<br />

0.79+<br />

0.32<br />

2.08+<br />

0.57 a<br />

2.13+<br />

1.08<br />

2.04+<br />

0.48 (a)<br />

2. 77+<br />

1.64<br />

1.87+<br />

0.22 (a)<br />

2.62+<br />

1.03 (a)<br />

2.85+<br />

1.17<br />

1.90+<br />

0.50<br />

1.51+<br />

0.20 (a)<br />

1.37+<br />

0.22<br />

6.82+<br />

0.45 b<br />

2.65+<br />

0.21 (a)<br />

2.08+<br />

0.16 b<br />

2.84+<br />

0.56 b<br />

1.73+<br />

0.32<br />

1.35+<br />

0.14<br />

1.23+<br />

0.14<br />

4.88+<br />

0.16 (a)<br />

3.09+<br />

0.40 b<br />

1.89+<br />

0.40<br />

1.62+<br />

0.10<br />

3.27+<br />

0.50 b<br />

2.25+<br />

0.28 b<br />

2.93+<br />

0.35 b<br />

7.62+<br />

6.33 (a)<br />

2.60+<br />

0.90 (a)<br />

3.38+<br />

0.81 b<br />

2.52+<br />

0.28 b<br />

1.99+<br />

0.58 a<br />

1.53+<br />

0.25 (a)<br />

1.64+<br />

0.38<br />

3.84+<br />

0.33 (a)<br />

2.21+<br />

0.32 (a)<br />

1.92+<br />

0.35 (a)<br />

2.30+<br />

0.50 (a)<br />

2.53+<br />

0.78 a<br />

1.55+<br />

0.30<br />

1.35+<br />

0.10<br />

5.65+<br />

1.63 (a)<br />

3.88+<br />

0.95 b<br />

2.52+<br />

0.50 (a)<br />

1.87+<br />

0.24<br />

144


Chapter 7: Manuscript V<br />

High-fat diet exposure is associated with renal carcinogenesis<br />

in rats<br />

Kerstin Stemmer 1* , Diego Perez-Tilve 2 , Daniel R. Dietrich 1 , Matthias H. Tschöp 2 , and Paul T.<br />

Pfluger 2<br />

1<br />

Human and Environmental Toxicology, University <strong>of</strong> Konstanz, Konstanz, Germany<br />

2<br />

Department <strong>of</strong> Psychiatry, Obesity Research Centre - Genome Research Institute, University<br />

<strong>of</strong> Cincinnati, College <strong>of</strong> Medicine, Cincinnati, Ohio, USA<br />

* Corresponding author: Kerstin Stemmer: kerstin.stemmer@uni-konstanz.de<br />

Manuscript to be submitted<br />

7.1 Abstract<br />

Animal and epidemiologic studies strongly suggest an association <strong>of</strong> renal cancer with dietinduced<br />

obesity. To date, however, it remains unclear whether the pro-carcinogenic<br />

processes in diet-induced obesity are a direct consequence <strong>of</strong> dietary lipds, or from an<br />

increase in adiposity with its co-morbidities and metabolic alterations. To elucidate the<br />

effects <strong>of</strong> high fat diet exposure and body adiposity on renal carcinogenesis, male Wistar<br />

rats were fed for 11 months with high fat diet, or chow. Life-history traits revealed two<br />

different rat subpopulations: one with a morbidly obese phenotype (Diet-Induced Obesity<br />

Sensitive, DIOsens, ca. 1.1kg bw), and one high-fat diet-resistant (DIOres) subgroup with<br />

modestly increased body weight (ca. 650g bw), and local fat depots more reminiscent <strong>of</strong><br />

chow-fed controls (ca. 470g bw).<br />

High fat-diet exposure led to a severe nephropathy, with early stages <strong>of</strong> renal<br />

carcinogenesis, such as atypical regenerative hyperplasia and preneoplasias in kidneys <strong>of</strong><br />

DIOsens and DIOres, and absence there<strong>of</strong> in control rats. In addition, increased cell<br />

proliferation was found in high-fat diet fed animals, and hyperphosphorylation <strong>of</strong> the S6<br />

ribosomal protein suggests an activated mTOR pathway in proliferating tubules in DIOsens<br />

and DIOres rats.<br />

145


Chapter 7: Manuscript V<br />

In conclusion, the similar degree <strong>of</strong> histopathological changes in DIOsens and DIOres rats<br />

indicates that the dietary fat content, rather than the degree <strong>of</strong> adiposity, is the key<br />

etiological factor for renal non-neoplastic and pre-neoplastic pathology. The here presented<br />

findings <strong>of</strong> increased mTOR activation in preneoplastic lesions <strong>of</strong> high fat diet-exposed rats<br />

independently <strong>of</strong> their degree <strong>of</strong> adiposity is consistent with a central role for this pathway<br />

in the pathogenesis <strong>of</strong> obesity induced carcinogenous lesions. With recurring cycles <strong>of</strong><br />

tissue damage, inflammation and regeneration are critical hallmarks <strong>of</strong> kidney cancer,<br />

chronic high-fat diet exposure may propagate the fixation and progression <strong>of</strong> spontaneous<br />

mutations and pre-neoplastic lesions.<br />

7.2 Introduction<br />

Obesity is defined as abnormal or excessive fat accumulation that presents a risk to health.<br />

It can arise from the interaction <strong>of</strong> both genetic factors and environmental conditions and<br />

behavior. A high availability <strong>of</strong> calorie-dense food, paired with a sedentary life style, thereby<br />

leads to a chronically positive energy balance with high caloric intake and low energy<br />

expenditure, and subsequently to the excessive storage <strong>of</strong> fat. It is assumed that<br />

differences in lifestyle, e.g. a lower spontaneous physical activity, or a lower metabolic rate<br />

(Ravussin and Danforth 1999; Ravussin 2005) are important factors influencing the<br />

relationship between high caloric intake body weight gain. Similar to human populations,<br />

inbreed rats also show varying susceptibilities to weight gain after high fat diet exposure<br />

(Levin et al. 1987), and are therefore useful tools to study the effects <strong>of</strong> high caloric or high<br />

fat exposure.<br />

In Western societies, the widespread obesity epidemia is accompanied by a concurrent<br />

increase in many accompanying diseases, such as diabetes, cardiovascular diseases and<br />

cancer (McTiernan 2005; Hotamisligil 2006; Van Gaal et al. 2006; Ogden et al. 2007). A<br />

recent survey <strong>of</strong> all available literature on adiposity and cancer (including epidemiologic,<br />

clinical, and experimental data) by the International Agency for Research on <strong>Cancer</strong><br />

(IARC), provided sufficient evidence that excess body weight increases the risk <strong>of</strong> cancers<br />

<strong>of</strong> the colon, breast (postmenopausal), endometrium, esophagus and the kidney (renal cell<br />

tumors) (IARC 2002a). Specifically, studies <strong>of</strong> populations worldwide suggested a 1.5- to<br />

2.5-fold higher relative risk <strong>of</strong> renal-cell cancer (RCC) for overweight and obese subjects<br />

compared to normal weight controls. Since kidney tumors are <strong>of</strong>ten only detected at late<br />

stages due to the absence <strong>of</strong> early symptoms, overall mortality rates are high. Worldwide,<br />

146


Chapter 7: Manuscript V<br />

kidney cancer with approximately 210.000 cases per year accounts for up to 2% <strong>of</strong> all<br />

malignancies.<br />

There is convincing pre-clinical and clinical evidence that renal cancer is being promoted or<br />

caused by high fat diet induced obesity (Mellemgaard et al. 1995; Chow et al. 2000;<br />

Bergstrom et al. 2001; Renehan et al. 2008), However, it is not clear whether renal cancer<br />

risk is increased by the high body adiposity itself with its manifold co-morbidities and<br />

disease consequences, such as subchronic inflammation, or non-physiological adipokine<br />

secretory patterns. Instead, the overload <strong>of</strong> dietary fat could directly elicit a negative effect<br />

on kidney physiology by direct stimulation <strong>of</strong> cellular signaling pathways through specific<br />

dietary fatty acids.<br />

One prime candidate for such a specific cellular effect could be the mammalian target <strong>of</strong><br />

rapamycin (mTOR) pathway, which plays a key role in both kidney cancer (Kenerson et al.<br />

2002; Albanell et al. 2007; Stemmer et al. 2007; Hanna et al. 2008) and obesity (Thomas<br />

2006). A very recent report suggested that high-fat diet exposure induced the activation <strong>of</strong><br />

the hypothalamic mTOR-S6K signaling pathway in rodents (Ono et al. 2008). Based on<br />

these findings, it was hypothesized in this study that high-fat diet exposure may induces<br />

renal mTOR-S6K activation, and thus may represent a key step in high-fat diet- induced<br />

renal tumorigenesis.<br />

The development <strong>of</strong> kidney tumors is frequently associated with chemically induced renal<br />

α2u-globulin accumulation in male rats, resulting in α2u-globulin nephropathy (Swenberg<br />

1993; Hard 1998; Doi et al. 2007). Since an early report identified α2u-globulin as kidney<br />

fatty acid binding protein (Kimura et al. 1989), it was further hypothesized that α2u-globulin<br />

might bind dietary lipids, might thereby get accumulated in rat kidneys after chronic high-fat<br />

diet exposure, and might thus result in renal pathology either directly or through an<br />

increased lipid accumulation in renal tubules.<br />

To elucidate the roles <strong>of</strong> chronic high-fat diet exposure and body adiposity on renal<br />

carcinogenesis, α2u-globulin accumulation and renal mTOR-S6K signaling, a novel rodent<br />

model <strong>of</strong> diet-induced obesity was employed. Specifically, diet-induced obesity sensitive<br />

(DIOsens) and diet-induced obesity resistant (DIOres) rats were selected from a large<br />

population <strong>of</strong> chronically high-fed diet fed Wistar rats, and life-history traits, plasma<br />

parameters, renal histopathology and cell proliferation were recorded and compared to<br />

those from a group <strong>of</strong> chow-fed animals. Thus, for the first time it was possible to dissect<br />

the direct influence <strong>of</strong> dietary fat vs. the degree <strong>of</strong> body adiposity on renal carcinogenesis.<br />

147


7.3 Material and Methods<br />

7.3.1 Animals<br />

Chapter 7: Manuscript V<br />

32 male Wistar rats (initial BW approx. 250g; Harlan, Indianapolis, IN, USA) were allowed<br />

ad libitum access to a customized high fat diet (Ref.: # D03082706, 40% butter fat, 46%<br />

carbohydrates (corn starch), 0.05% cholesterol, Research Diets, New Brunswick, NJ) for 11<br />

months. From the initial cohort, two subpopulations <strong>of</strong> 1) diet-induced obesity resistant rats<br />

(DR) and 2) diet-induced obesity sensitive rats (DIO) were studied further in comparison to<br />

a group <strong>of</strong> standard chow-fed rats (6% fat, Harlan Teklad LM-485).<br />

All rats were housed in individual cages and maintained on a 12 hr light – dark cycle in a<br />

temperature controlled environment at 23°C. Ad libitum fed animals were sacrificed by<br />

decapitation, and the kidneys rapidly excised. <strong>Kidney</strong>s were cross sectioned and sections<br />

were either frozen on dry ice and kept at -80˚C, or fixed in 10% buffered formalin for a total<br />

<strong>of</strong> 4 months before paraffin embedding. In addition, epididymal fat pads were excised and<br />

weighed. All procedures were approved by the Institutional Animal Care and Use<br />

Committees at the University <strong>of</strong> Cincinnati Office in accordance with the NIH guide for the<br />

care and use <strong>of</strong> laboratory animals.<br />

7.3.2 Blood parameters<br />

Two weeks prior to sacrifice, tail blood was collected from overnight fasted rats,<br />

immediately chilled on ice, and centrifuged at 2500g, 4°C for 10min to extract plasma.<br />

Circulating non-esterified fatty acid, triglyceride and cholesterol levels were determined<br />

using commercially available enzymatic assays (NEFA, Wako Germany; Infinity triglyceride<br />

and Infinity cholesterol, Thermo Electron). Circulating adipokine levels were measured by<br />

using the respective multiplex mouse panels from LINCOplex (St. Charles, MO, USA). All<br />

measurements were performed in duplicate according to the manufacturer’s instructions.<br />

Fed and fasting blood glucose levels were measured directly from tail blood by using a<br />

glucometer (TheraSense Freestyle).<br />

148


7.3.3 Renal Pathology<br />

Chapter 7: Manuscript V<br />

H&E stained paraffin sections were randomized for histopathological evaluation, Nonneoplastic<br />

pathology was classified as none (0), mild (1), moderate (2), strong (3), and<br />

severe (4). Total numbers <strong>of</strong> preneoplastic lesions were counted and depicted as<br />

incidences/section & animal. All pathological examinations were conducted by KS and<br />

independently verified by DRD.<br />

7.3.4 Cell Proliferation analysis<br />

Cell proliferation was evaluated by immunohistochemical staining for proliferating cell<br />

nuclear antigen (PCNA) using monoclonal primary anti-PCNA antibody, in paraffinembedded<br />

kidney sections, as described previously (Stemmer et al. 2007). For details see<br />

table 7.1. PCNA- positive stained S-phase nuclei were quantified on randomized sections.<br />

20 microscopic fields (10x ocular, 40x objective) were randomly chosen within the area <strong>of</strong><br />

the renal outer cortex and inner cortex/outer medulla. A minimum <strong>of</strong> 1000 proximal tubules<br />

were counted separately per field, distinguishing between negative and positive PCNAstained<br />

nuclei. Nuclear labeling indices (LI) were calculated as positive nuclei/total number<br />

<strong>of</strong> nuclei counted.<br />

Table 7.1: Overview <strong>of</strong> antibodies and modifications to the manufacture’s protocols<br />

Primary antibody Company Antibody retrival Dilution Incubation<br />

Mouse Anti- PCNA<br />

(PC-10)<br />

Rabbit anti-pS6RP<br />

(Ser235/236)<br />

Mouse anti-alpha2uglobulin<br />

DAKO, Cat No:<br />

M0879<br />

Cell Signaling<br />

Technologies, Cat<br />

No: 2211<br />

Kind gift from Dr. U.<br />

Deschel (Boehringer<br />

Ingelheim, Biberach,<br />

Germany)<br />

7.3.5 Immunohistochemistry<br />

10mM sodium<br />

citrate buffer (pH<br />

6.0), 10min at 98°C<br />

10mM sodium<br />

citrate buffer (pH<br />

6.0), 10min at 98°C<br />

1: 50 in 1x<br />

powerblock:<br />

Biogenex; Cat No.<br />

HK085-5K<br />

1:100 in 5% normal<br />

goat serum<br />

none 1: 200 in 1x<br />

powerblock:<br />

Biogenex; Cat No.<br />

HK085-5K<br />

Over night at<br />

4°C<br />

Over night at<br />

4°C<br />

Over night at<br />

4°C<br />

Immunohistochemistry <strong>of</strong> phospho-S6 ribosomal protein (pS6RP), PCNA, and α2u-globulin<br />

in paraffin sections was carried out according to the manufacturers’ instructions (for brief<br />

information see table 7.1). Antigen–antibody complexes were visualized using the Super<br />

149


Chapter 7: Manuscript V<br />

SensitiveTM (BioGenex, USA) alkaline phosphatase-labeled, biotin–streptavidin amplified<br />

detection system and Fast Red as chromogen.<br />

7.3.6 Statistics<br />

Significant differences in nuclear labeling indices (LI%), or in the total number <strong>of</strong><br />

preneoplastic lesions, were calculated by using Bonferroni's test for multiple comparisons.<br />

Ranked non-neoplastic pathology data were analyzed by using Kruskal-Wallis tests. All<br />

analyses were performed by using GraphPad Prism 5.0 (GraphPad S<strong>of</strong>tware, San Diego,<br />

California, USA). All results are given as Means ± SD. Results were considered statistically<br />

significant when p < 0.05 (*p < 0.05; **p < 0.01 and ***p


Chapter 7: Manuscript V<br />

Table 7.2: Life-history traits <strong>of</strong> chow-fed rats (n=8), and DIOres (n=8) and DIOsens (n=7)<br />

subpopulations exposed to high fat diet.<br />

Body weight [g]<br />

Epididymal fat pad weights<br />

[g]<br />

Fasting glucose [mg/dL]<br />

Fed glucose [mg/dL]<br />

Triglyceride<br />

Free fatty acids<br />

Chow DIOres DIOsens<br />

468.0±10.9 649.6±15.9 1031.1±34.8<br />

20,5±1.3 76.5±7.9 204.4±12.5<br />

72.63±1.7 77.1±1.9 85.1±3.1<br />

99.6±2.4 99.8±3.7 104.5±3.0<br />

192.6±15.8 243.6±72.8 244.6±46.7<br />

0.44±0.030 0.41±0.033 0.37±0.027<br />

Despite the marked changes in body weight and fat mass in DIOsens and DIOres<br />

compared to chow-fed rats, no changes were observed in total fasting triglyceride,<br />

cholesterol or free fatty acid levels between the groups (Table 7.2).<br />

7.4.2 Increased levels <strong>of</strong> adipokines and insulin in DIOsens and DIOres<br />

rats<br />

Leptin levels were increased in both DIOsens and DIOres rats, compared to chow fed<br />

controls, however, the degree <strong>of</strong> body adiposity did not influence leptin levels in DIOres vs.<br />

DIOsens rats (Figure 7.1, A). In contrast, only DIOsens rats had higher insulin levels,<br />

compared to chow-fed or DIOres rats (Figure 7.1, B). Plasminogen activator inhibitor 1<br />

(PAI-1) levels correlated to the degree <strong>of</strong> body adiposity, and were higher in DIOres and<br />

especially in DIOsens rats (Figure 7.1, E). No changes were observed for Interleukin 1β (IL-<br />

1β) or monocyte chemoattractant protein 1 (MCP-1), or in any <strong>of</strong> the groups tested (Figure<br />

7.1, C and D).<br />

151


Chapter 7: Manuscript V<br />

Figure 7.1: Adipokine, cytokine, and insulin levels in plasma from chow-fed controls (open bars,<br />

n=8), and high-fat diet fed DIOres (striped bars, n=8), and DIOsens (grey bars, n=7)<br />

rats. All rats were fed ad libitum prior to blood sampling. Significant differences<br />

between groups were evaluated by using one-way ANOVA and Bonferroni's Post Test<br />

for multiple comparisons.<br />

7.4.3 High-fat diet exposure causes preneoplasia in DIOsens and DIOres<br />

rats<br />

Chronic high fat diet exposure resulted in an increased non-neoplastic renal pathology only<br />

in DIOsens and DIOres rats, but not in rats fed a control diet (Table 7.3). Most prominent<br />

changes included a marked chronic progressive nephropathy (CPN) <strong>of</strong> the renal cortex,<br />

including tubular degeneration and regeneration, glomerulosclerosis, interstitial fibrosis and<br />

tubules containing protein casts and a widespread mononuclear cell inflammation (Figure<br />

7.2, A and B). Notably, CPN in DIOsens and DIOres rats was associated with simple<br />

tubular hyperplasia characterized by an increased number <strong>of</strong> epithelial cells that do not<br />

extend beyond the single cell layer (Figure 7.2, A and B). Importantly, some tubules within<br />

the CPN lesions had a preneoplastic phenotype: partly solid, with basophilic cytoplasm,<br />

irregular shaped nuclei and not associated with a markedly thickened basement membrane<br />

(Figure 7.2, C and D). Notably, no overt changes in any <strong>of</strong> the observed parameters were<br />

observed between DIOres and DIOsens rats.<br />

152


Chapter 7: Manuscript V<br />

Table 7.3: Histopathology in chow-fed (n=8), DIOres (n=8) and DIOsens (n=7) rats exposed to<br />

high fat diet. Histopathological changes were ranked from none (0) to severe (4)<br />

including intermediate classes. Values are presented as median ± median absolute<br />

deviation.<br />

Glomeruli<br />

Cortex/Medulla<br />

Chow DIOres DIOsens<br />

Glomerulosclerosis 0.0 + 0.0 0.8 + 0.3 1.0 + 0.3*<br />

Fibrosis 0.0 + 0.0 0.8 + 0.3 0.5 + 0.2<br />

Dilated blood vessels 0.0 + 0.0 0.5 + 0.3 0.0 + 0.2<br />

Proteincast in Bowman’s<br />

space<br />

0.0 + 0.0 0.0 + 0.2 0.0 + 0.1<br />

Necrosis 0.0 + 0.1 0.5 + 0.4 1.0 + 0.5<br />

Vacuolisation 0.0 + 0.0 0.0 + 0.5 0.0 + 0.5<br />

Apoptosis 0.0 + 0.0 0.0 + 0.0 0.0 + 0.3<br />

CPN 0.3 + 0.3 1.5 + 0.6* 2.0 + 0.5***<br />

Regeneration 0.3 + 0.3 1.0 + 0.5* 1.5 + 0.3*<br />

Regenerative Hyperplasia 0.0 + 0.0 0.8 + 0.6 1.0 + 0.2<br />

Inflammation 0.0 + 0.2 1.3 + 0.6* 2.0 + 0.6**<br />

Protein Casts 0.0 + 0.1 1.3 + 0.6** 1.5 + 0.7***<br />

Calcium Casts 0.0 + 0.1 0.0 + 0.2 0.5 + 0.2<br />

Immunhistochmenistry alpha2u-globulin 2.5 + 0.1 2.5 + 0.4 2.5 + 0.1<br />

* **<br />

Significantly higher than the respective control group. Kruskal-Wallis test (p< 0.05). Significantly higher<br />

than the respective control group. Kruskal-Wallis test (p< 0.005). *** Significantly higher than the respective<br />

control group. Kruskal-Wallis test (p< 0.001)<br />

153


Chapter 7: Manuscript V<br />

Figure 7.2: Representative images <strong>of</strong> (A, B) simple tubular hyperplasia with tubules containing a<br />

thickened basal membrane and increased number <strong>of</strong> epithelial cells that do not<br />

extend beyond the single cell layer (1). Images <strong>of</strong> atypical hyperplasia (C, D), partly<br />

solid, with basophilic cytoplasm, irregular shaped nuclei and not associated with a<br />

markedly thickened membrane (2). Protein cast (3). Representative alpha 2u stained<br />

protein droplets in DIOsens rats (E, F).<br />

154


Chapter 7: Manuscript V<br />

7.4.4 High-fat diet-induced changes in kidney pathology are<br />

independent from α2u-globulin accumulation<br />

Positively stained cytoplasmatic α2u-globulin droplets were localized within the P2 segment<br />

<strong>of</strong> the proximal convoluted tubule. When classifying α2u-globulin staining intensity as none<br />

(0), mild (1), moderate (2), strong (3), and severe (4), similar intensities and therefore<br />

amounts <strong>of</strong> α2u-globulin were found in all groups tested, independent <strong>of</strong> the diet and<br />

severity <strong>of</strong> obesity (see last row in table 7.3, and Figure 7.2, E and F). In addition, no<br />

difference in the tubular localisation was detected, compared to the control rats. Thus, α2u-<br />

globulin did not accumulate with high-fat diet exposure, and did not seem to be implicated<br />

in the observed changes in renal pathology.<br />

7.4.5 High fat diet increases cell proliferation in proximal tubule <strong>of</strong><br />

DIOsens and DIOres rats<br />

Visualization <strong>of</strong> S-Phase nuclei by immunohistochemical staining <strong>of</strong> nuclear PCNA<br />

demonstrated a marked increase <strong>of</strong> proliferating cells within the CPN lesions compared to<br />

the healthy tissue. To determine whether high fat diet targets distinct tubular segments, cell<br />

proliferation was separately analyzed in random fields in specifiable areas <strong>of</strong> the outer and<br />

inner cortex. This resulted in the detection <strong>of</strong> a similar and significant increase <strong>of</strong> cell<br />

proliferation in the p1/p2 segments and p3 segments proximal tubules <strong>of</strong> DIOsens and<br />

DIOres rats respectively, compared to chow fed control rats (Figure 7.3, A and B).<br />

155


Chapter 7: Manuscript V<br />

Figure 7.3: (A) Representative images <strong>of</strong> PCNA staining in DIOsens rats. Red: PCNA positive<br />

nuclei, blue, negative counterstaining with haematoxilin. (B) Comparison <strong>of</strong> PCNA<br />

labelling indices (LI%) <strong>of</strong> Eker rats <strong>of</strong> chow fed, DIOsens and DIOres rats exposed<br />

with high fat diet. LI%, determined for proximal tubules (PT) within randomly chosen<br />

fields <strong>of</strong> the outer or inner cortex. Within both regions at least 1000 nuclei were<br />

counted. LI% were tested for significance using one-way ANOVA and Bonferroni's<br />

Post Test for multiple comparisons.<br />

7.4.6 Involvement <strong>of</strong> the mTOR pathway<br />

To evaluate the potential activation <strong>of</strong> the mTOR-S6K pathway by high-fat diet exposure,<br />

phosphorylation <strong>of</strong> Ser235/236 residues <strong>of</strong> S6RP was measured by immunohistochemical<br />

detection. S6RP Ser235/236 specifically gets phosphorylated by activated S6K1 (p70<br />

ribosomal protein S6 kinase 1) (Flotow and Thomas 1992) and serves as marker for<br />

mTOR-S6K activation. Only single S6RP phosphorylation positive cells were found in<br />

chow-fed control rats, or in unaffected tissue <strong>of</strong> any group (Figure 7.4, A). In contrast, in<br />

both DIOsens and DIOres rats positive staining was detected in nearly all cells <strong>of</strong><br />

regenerating tubules within the CPN lesions, and especially in tubules with an atypical<br />

156


Chapter 7: Manuscript V<br />

phenotype (Figure 7.4, B-D). However, no overt changes in S6RP phoshphorylation could<br />

be observed between DIOsens and DIOres rats.<br />

Figure 7.4: Representative images <strong>of</strong> single cell pS6RP staining (1) <strong>of</strong> a chow fed control rats,<br />

proliferating pS6RP positive tubules with in DIOsens rat (B-D). pS6RP positive tubular<br />

cell undergoing mitosis (2).<br />

7.5 Discussion<br />

Renal tissue damage, inflammation and regeneration are important factors contributing to<br />

the onset and progession <strong>of</strong> RCC by propagating the fixation <strong>of</strong> spontaneous mutations and<br />

progression <strong>of</strong> pre-neoplastic lesions into solid renal tumors (Dietrich and Swenberg 1990;<br />

Cohen and Ellwein 1991; Cohen and Arnold 2008). Obesity is a known risk factor for kidney<br />

cancer, and chronic exposure to dietary lipids has previously been shown to cause<br />

inflammation, regeneration and renal pathologies in animal models for obesity (Aguila and<br />

Mandarim-De-Lacerda 2003; Armitage et al. 2005; Altunkaynak et al. 2008). However,<br />

157


Chapter 7: Manuscript V<br />

none <strong>of</strong> these previous studies could convincingly distinguish between the direct influence<br />

<strong>of</strong> dietary fat, and the impact <strong>of</strong> the high body adiposity.<br />

This study describes for the first time the effect <strong>of</strong> chronic high fat diet exposure on renal<br />

pathology in DIOsens and DIOres rats. These rats were selected from a large population <strong>of</strong><br />

high-fat diet fed inbred Wistar rats, and showed marked differences in their body weight<br />

and fat pad weight despite the highly similar genetic background. High-fat diet exposure<br />

induced marked chronic progressive nephropathy (CPN) and an increased regenerative<br />

cell proliferation <strong>of</strong> similar severity in both DIOsens and DIOres rats, compared to chow fed<br />

controls. Notably, in DIOsens and DIOres rats, CPN was not only associated with a simple<br />

tubular hyperplasia, but also with preneoplastic lesions. Although neither DIOsens nor<br />

DIOres rats presented with full adenomas or carcinomas after high fat diet exposure, it is<br />

now widely accepted that atypical hyperplasias represent a preneoplastic entity which has<br />

the potential to progress to adenomas and carcinomas (Dietrich and Swenberg 1991; Hard<br />

and Khan 2004). However, it remains unclear whether atypical tubular hyperplasias, or<br />

other renal pathologies, are the result <strong>of</strong> high adiposity with its direct physiological<br />

consequences, <strong>of</strong> co-morbidities <strong>of</strong> obesity such as hypertension or diabetes, or <strong>of</strong> direct<br />

toxic effects <strong>of</strong> dietary fat.<br />

7.5.1 Similar renal preneoplasias in DIOsens and DIOres rats suggest a<br />

minor role <strong>of</strong> high body adiposity or its co-morbidities<br />

The lack <strong>of</strong> overt differences in non-neoplastic pathology between DIOres and DIOsens<br />

rats, points to a minor role for the level <strong>of</strong> adiposity in the progression <strong>of</strong> non-neoplastic<br />

pathology. However, despite significantly higher epididymal fat pad weights, leptin levels in<br />

DIOsens rats were only slightly higher than in DIOres rats. Previously, an excess secretion<br />

<strong>of</strong> leptin in obese animals was suggested to mediate, at least in part, the effects <strong>of</strong> high<br />

body adiposity on renal carcinogenesis (Takahashi et al. 1996; Attoub et al. 2000;<br />

Horiguchi et al. 2006). Thus, leptin might in part contribute to the observed renal pathology<br />

in DIOsens and DIOres rats. In obesity, monocyte infiltration <strong>of</strong> white adipose tissue can<br />

lead to the release <strong>of</strong> cytokines, and a sub-chronic, systemic inflammation. However,<br />

similar plasma levels <strong>of</strong> MCP1 and IL-ß in all groups tested do not suggest such an<br />

increased cytokine release, or sub-chronic inflammation. PAI-1 levels were reported to be<br />

elevated in obesity and might enhance fibrinolysis in renal disease by inhibiting plasmindependent<br />

extracellular matrix turnover, thus enhancing renal pathology (Eddy and Fogo<br />

2006). Accordingly, PAI-1 levels increased with the degree <strong>of</strong> body adiposity in DIOres rats<br />

158


Chapter 7: Manuscript V<br />

and DIOsens rats, compared to chow fed controls. Adipokine levels in diet-induced obesity<br />

might therefore contribute in part to the observed effects, however, their exact role in the<br />

etiology <strong>of</strong> kidney cancer still remains elusive. For instance, A-Zip/F-1 mice, which<br />

completely lack white adipose tissue and therefore have undetectable adipokine levels,<br />

nevertheless displayed higher tumor incidences, tumor multiplicity, and decreased tumor<br />

latency than wild-type mice in two experimental tumor models (Nunez et al. 2006).<br />

Insulin resistance, diabetes type II and associated hyperglycemia itself are well-established<br />

risk factors for the onset <strong>of</strong> renal cancer (Hjartaker et al. 2008). Chronic hyperinsulinemia<br />

may thereby induce an increased synthesis and bioavailability <strong>of</strong> the insulin-like growth<br />

factor 1 (IGF1), one <strong>of</strong> the most potent activators <strong>of</strong> the oncogenic AKT signaling pathway<br />

(Renehan et al. 2006). Hyperglycemia may further induce the localized production <strong>of</strong><br />

reactive oxygen species (ROS) via multiple mechanisms such as advanced glycation or<br />

enhanced glycolysis, and may ultimately lead to diabetic nephropathy (Coughlan et al.<br />

2008; Forbes et al. 2008) and renal DNA damage (Sebekova et al. 2007), therefore<br />

increasing the risk for kidney cancer. However, despite severe adiposity especially in the<br />

DIOS rats, similar blood glucose levels (ad libitum and fasted) in DIOres, DIOsens and<br />

chow fed animals indicated that high fat diet exposure did not induce hyperglycemia, one<br />

major hallmark <strong>of</strong> type II diabetes. In addition, insulin levels were only increased in<br />

DIOsens, but not in DIOres rats, compared to chow-fed controls. Based on the similar renal<br />

pathology <strong>of</strong> DIOsens and DIOres rats, hyperglycemia and hyperinsulinemia therefore do<br />

not seem to induce the here observed changes. Thus, the here presented findings suggest<br />

that the influence <strong>of</strong> body adiposity on renal pathology and renal carcinogenesis is rather<br />

minor.<br />

7.5.2 High-fat diet exposure does not induce α2u-globulin accumulation<br />

Dietary lipids may have a direct effect on renal pathology and early markers <strong>of</strong> renal<br />

tumorigenesis. It was thus initially hypothesized that an excessive renal accumulation <strong>of</strong> the<br />

fatty acid binding protein α2u-globulin may contribute to high fat diet-induced CPN, but<br />

immunohistochemical staining <strong>of</strong> α2u-globulin could not corroborate that hypothesis.<br />

However, glomerusclerosis, and tubular damage mainly within the proximal part <strong>of</strong> the renal<br />

nephron in DIOsens and DIOres rats might point to a direct toxic effect <strong>of</strong> lipids. Such<br />

effects were previously observed after chronic high fat diet feeding in leptin-receptor<br />

deficient db/db mice, or diet-induced obese C57BL/6J mice, and were attributed in part<br />

towards a sterol regulatory element binding protein (SREBP)-mediated lipid accumulation<br />

159


Chapter 7: Manuscript V<br />

in the kidney (Jiang et al. 2005; Wang et al. 2005). Previous reports suggested a different<br />

capacity for lipid metabolism within different tubular segments, with mitochondrial betaoxidation<br />

<strong>of</strong> fatty acids being relatively small in the proximal and the distal nephron, but<br />

high in the proximal tubule. Notably, the proximal tubule has a peroxisomal beta-oxidative<br />

capacity <strong>of</strong> the same order <strong>of</strong> magnitude as in liver cells <strong>of</strong> Wistar rats (Le Hir and Dubach<br />

1982). Higher lipid turnover in the glomerulus or proximal tubule may therefore lead to the<br />

release <strong>of</strong> reactive oxygen species by mitochondria, and regional oxidative stress, one key<br />

player in the pathogenesis <strong>of</strong> renal pathology.<br />

7.5.3 mTor-S6K signaling: The link between obesity and kidney cancer<br />

<strong>Kidney</strong> cancer has been linked to an abnormal activation <strong>of</strong> the mammalian target <strong>of</strong><br />

rapamycin (mTOR) pathway (Kenerson et al. 2002; Albanell et al. 2007; Stemmer et al.<br />

2007; Hanna et al. 2008). Activation <strong>of</strong> the mTOR pathway results in activation <strong>of</strong> its<br />

downstream target S6-Kinase (S6K) and further to phosphorylation <strong>of</strong> S6 ribosomal protein<br />

(S6RP) at Ser293. Due to the lack <strong>of</strong> S6K antibodies suitable for immunohistochemistry,<br />

staining <strong>of</strong> phosphoSer293- S6RP was used as surrogate marker for an activated mTOR<br />

pathway. The here presented data clearly demonstrate that mTOR pathway activation can<br />

only be observed in DIOsens and DIOres rats. Specifically, mTOR signaling is activated<br />

specifically in proximal tubules that also demonstrate an increased proliferation and<br />

regenerative hyperplasia with atypical nuclear morphology. The functional role <strong>of</strong> such<br />

mTor/S6K pathway activation in tumor initiation and/or tumor promotion, however, has not<br />

been clearly established. Kenerson and co-workers demonstrated that rapamycin, an<br />

inhibitor <strong>of</strong> mTOR, diminished renal preneoplastic progression to veritable renal tumors in<br />

Eker rats but not the formation <strong>of</strong> earliest preneoplastic lesions (Kenerson et al. 2002). In<br />

addition, an activation <strong>of</strong> the mTOR/S6K pathway might serve as facilitator <strong>of</strong> tumor<br />

initiation by an induction <strong>of</strong> cell proliferation, thus elevating the risk for the fixation <strong>of</strong> DNA<br />

mutations, e.g. induced by increased oxidative stress.<br />

The mTOR pathway is also known to be a central regulator for the nutrient-hormonal<br />

signaling network (Cota et al. 2008; Gulati et al. 2008), and deregulation by high-fat diet<br />

exposure might therefore induce human and rodent cancer. High fat diet exposure was<br />

previously shown to induce hypothalamic S6K activation (Ono et al. 2008). In addition,<br />

Palmitoleic acid was shown to rapidly induce S6K phosphorylation in various hypothalamic<br />

and muscle cell lines in vitro, and chronic exposure <strong>of</strong> mice to high-fat diet induced the<br />

phosphorylation <strong>of</strong> skeletal muscle S6K, whereas exposure to a fat-free diet reduced<br />

160


Chapter 7: Manuscript V<br />

skeletal muscle S6K phosphorylation (Castaneda et al. 2008). Thus, it seems entirely<br />

possible that fatty acids, derived from high-fat diet, could also activate S6K in renal cells.<br />

Further studies, based on in vitro exposure <strong>of</strong> primary kidney cells or established kidney<br />

cell lines with various fatty acids, will help to clarify this open question.<br />

7.5.4 Conclusion<br />

The here presented data suggests that dietary lipids can induce renal toxicity. This effect<br />

seems largely independent <strong>of</strong> the level <strong>of</strong> adiposity, and can manifest in non-diabetic rats.<br />

The high-fat diet induced activation <strong>of</strong> the mTOR pathway by dietary lipids with the<br />

increase in cell proliferation, may promote the fixation and progression <strong>of</strong> spontaneous<br />

mutations and the development <strong>of</strong> neoplasia and solid tumors, and may causally link<br />

obesity and renal cancer.<br />

161


Chapter 8: Additional Data<br />

8.1 Gender Disparity in Early Ochratoxin A and Aristolochic<br />

Acid Mediated Renal Toxicity in Short-term Assays<br />

This chapter summarizes additional data from short-term AA- and OTA-treated female Eker<br />

and wild type rats, respectively, that were not presented in one <strong>of</strong> the previous manuscripts.<br />

The experiments and analyses in female rats were done together with those from male<br />

rats, which are shown in chapter 3. Therefore, sex-specific effects <strong>of</strong> AA or OTA treatment<br />

on cell proliferation and renal pathology in female rats can be directly compared to those<br />

derived from male rats. For details on the experimental design, see material and methods<br />

<strong>of</strong> chapter 3. The results from these gender disparity experiments were partly presented as<br />

poster at the 47 th Annual Meeting <strong>of</strong> the Society <strong>of</strong> Toxicology, 2008 in Seattle, WA, USA.<br />

8.2 Ochratoxin A<br />

8.2.1 Cell proliferation (PCNA labeling index)<br />

Treatment <strong>of</strong> male and female Eker and wild typ rats with 210µg OTA/kg bw significantly<br />

increased cell proliferation in the renal cortex after 7 and 14 days <strong>of</strong> exposure (Figure 8.1).<br />

Male rats showed a strong tendency towards a higher OTA-dependent increase in cell<br />

proliferation, compared to female rats. Furthermore, the gender difference in cell<br />

proliferation was more pronounced in wild type than in Eker rats.<br />

162


Chapter 8: Additional Data<br />

Figure 8.1: Comparison <strong>of</strong> PCNA labelling indices (LI%) in the renal cortex <strong>of</strong> OTA-treated Eker<br />

and wild type rats. Data represent the mean + SD <strong>of</strong> n=3 animals per group.<br />

Significant differences between treated and time-matched control groups: *p < 0.05;<br />

**p < 0.01 and ***p


Chapter 8: Additional Data<br />

Figure 8.2: Comparison <strong>of</strong> total numbers and incidences <strong>of</strong> basophilic atypical tubules (bATs) in<br />

male and female Eker and wild type rats treated with OTA. Significant differences<br />

between treated and time-matched control groups: *p < 0.05; **p < 0.01 and ***p<br />


8.3 Aristolochic acid<br />

8.3.1 Cell proliferation (PCNA labeling index)<br />

Chapter 8: Additional Data<br />

Treatment <strong>of</strong> male and female Eker and wild typ rats with 10mg AA/kg bw significantly<br />

increased cell proliferation in the renal cortex <strong>of</strong> female but not male rats (Figure 8.3).<br />

Female Eker rats appeared to be more sensitive to AA than corresponding wild type rats,<br />

as an increased cell proliferation rate was already observed at day 7 <strong>of</strong> exposure.<br />

Figure 8.3: Comparison <strong>of</strong> PCNA labelling indices (LI%) in the renal cortex <strong>of</strong> AA-treated Eker<br />

and wild type rats. Data represent the mean + SD <strong>of</strong> n=3 animals per group.<br />

Significant differences between treated and time-matched control groups: *p < 0.05;<br />

**p < 0.01 and ***p


Chapter 8: Additional Data<br />

significantly increase the number <strong>of</strong> bATs in comparison to vehicle controls. Higher<br />

background numbers and incidences <strong>of</strong> bATs were found in TSC2-mutant Eker rats.<br />

Figure 8.4: Comparison <strong>of</strong> total numbers and incidences <strong>of</strong> basophilic atypical tubules (bATs) in<br />

male and female Eker and wild type rats treated with AA. Mean + SD. No significant<br />

differences between treated and time-matched control groups were detected by one<br />

way ANOVA with Bonferroni post-test.<br />

8.3.3 Conclusion<br />

Although no sex-differences in total numbers and incidences <strong>of</strong> preneoplastic lesions could<br />

be detected, the increased cell proliferation in AA-treated females but not in males suggest<br />

a higher sensitivity <strong>of</strong> female rats towards AA treatment. This is corroborated by previous<br />

findings, demonstrating 2-fold higher levels <strong>of</strong> dG-AAI and dA-AAII DNA adducts in kidneys<br />

<strong>of</strong> female than male Wistar rats following treatment with oral doses <strong>of</strong> 0.15mg/kg body<br />

weight for 3 months (Arlt et al. 2001).<br />

166


Chapter 9: Overall Discussion and Future Perspectives<br />

9.1 Short-term In Vivo Tests to Identify Renal Carcinogens<br />

Exposure to renal carcinogens remains one important cause for a vast majority <strong>of</strong> all<br />

human kidney cancers and identification <strong>of</strong> potential carcinogens serves as first step in<br />

disease prevention. Classification as carcinogen is mainly based on life-time rodent<br />

bioassays. These tests assume that cancer development is a basic process with a similar<br />

pathogenesis in humans and in experimental animals (Bannasch 1986a; Bannasch 1986b).<br />

However, the scientific value <strong>of</strong> rodent bioassays has come under criticism (Monro 1993;<br />

Davies et al. 2000; Haseman et al. 2001). The criticisms are: a) that the studies are too<br />

long and should rather be terminated at earlier points <strong>of</strong> life when the animals still have<br />

essentially normal organ functions and are free from widespread age-related pathology, b)<br />

that the carcinogenic doses are too high and, due to cytotoxicity and regenerative cell<br />

proliferation or dose-dependent differences in metabolism and pharmacokinetics, do not<br />

reflect the true carcinogenic potential, and c) that some organ-specific carcinogenic effects<br />

observed in experimental animals have little or no relevance to humans (e.g. α2u-globulin<br />

nephropathy associated with renal cancer in male rats, or chemically induced peroxisomal<br />

proliferation associated with rodent liver tumors). Other criticisms are that rodent bioassays<br />

require hundreds <strong>of</strong> animals, are time-consuming, and costly, especially due to the high<br />

amounts <strong>of</strong> compounds required.<br />

Due to the current limitations <strong>of</strong> the lifetime rodent bioassay, interest in developing sensitive<br />

short-term, mechanistically based in vivo studies has grown. One promising approach is<br />

the use <strong>of</strong> microarrays for the identification and characterization <strong>of</strong> novel mechanistic<br />

biomarkers involved in the early onset <strong>of</strong> cancer. In addition, since microarrays are highly<br />

sensitive detection tools, gene expression pr<strong>of</strong>iling may allow an early detection <strong>of</strong> adverse<br />

effects <strong>of</strong> test compounds, even before those effects have manifested in histopathological<br />

changes. However, before short-term assays in combination with gene expression pr<strong>of</strong>iling<br />

can be adopted as alternatives for the lifetime rodent bioassay, further research is needed<br />

to establish and validate their sensitivity, accuracy and predictivity to detect carcinogenic<br />

compounds. Some promising studies have been performed where gene expression pr<strong>of</strong>iles<br />

from rat livers were used to identify and distinguish known genotoxic and non-genotoxic<br />

167


Chapter 9: Overall Discussion and Future Perspectives<br />

hepato-carcinogens after 1 to 14 days <strong>of</strong> treatment (Ellinger-Ziegelbauer et al. 2005;<br />

Ellinger-Ziegelbauer et al. 2008). However, at the beginning <strong>of</strong> this thesis no comparable<br />

studies on renal carcinogens had been performed.<br />

The first aim <strong>of</strong> this thesis was therefore to investigate some <strong>of</strong> the most important<br />

prerequisites needed for a sensitive, specific and predictive short-term in vivo test to<br />

identify renal carcinogens. Those are:<br />

1. Detectable and compound-specific gene expression changes in kidney<br />

homogenate <strong>of</strong> short-term treated rats (Sensitivity)<br />

2. Possible differentiation between genotoxic and non-genotoxic renal carcinogens by<br />

these gene expression pr<strong>of</strong>iles (Specificity)<br />

3. Manifestation <strong>of</strong> early genotoxic or non-genotoxic expression changes in renal<br />

preneoplastic and neoplastic lesions (Predictivity)<br />

An in-depth discussion on the respective hypotheses and obtained results can be found in<br />

the respective manuscripts. This chapter will summarize the findings <strong>of</strong> all short-term and<br />

long-term in vivo experiments, and will discuss them in context to the above named<br />

prerequisites. Thus, a more comprehensive evaluation <strong>of</strong> all findings presented in this<br />

thesis might help to clarify whether the methods and model systems used for these studies<br />

can help to establish a new standard for renal toxicity testing. Ultimately, such a new<br />

standard test could shorten the duration <strong>of</strong> the life-time rodent bioassay, and could help to<br />

achieve the goal <strong>of</strong> the three R’s, to reduce, refine and replace animal experiments.<br />

9.1.1 Prerequisite I: Short term treatment would result in detectable<br />

compound-specific gene expression changes<br />

In the first part <strong>of</strong> this thesis, gene expression pr<strong>of</strong>iles <strong>of</strong> kidney cortex homogenates <strong>of</strong><br />

male Tsc2 mutant Eker- and corresponding wild-type rats were analyzed after 1, 3, 7 and<br />

14 days <strong>of</strong> treatment with genotoxic (AA and MAMAc) and non-genotoxic (OTA) renal<br />

carcinogens and compared with compound-induced changes in renal pathology and cell<br />

proliferation (for details see chapter 3, 4 and 8). In both Eker- and wild-type rats short-term<br />

treatment with the respective carcinogens resulted in minor but compound-specific changes<br />

in histopathology and cell proliferation that were reflected by early gene expression pr<strong>of</strong>iles.<br />

168


Chapter 9: Overall Discussion and Future Perspectives<br />

1. AA and OTA resulted in an up-regulation <strong>of</strong> genes and pathways involved in<br />

inflammatory responses, which was in consistence with the histopathological<br />

finding <strong>of</strong> an infiltration <strong>of</strong> monocytes in both groups.<br />

2. OTA induced the deregulation <strong>of</strong> genes modulated by acute toxic effects (e.g.<br />

genes involved in oxidative stress response, regenerative cell proliferation, and<br />

increased extracellular matrix synthesis). These changes correlated well with the<br />

histopathological findings in OTA-treated rats demonstrating tubular damage and<br />

regeneration with signs <strong>of</strong> fibrosis.<br />

3. OTA but not AA treatment resulted in an up-regulation <strong>of</strong> numerous genes involved<br />

in cell cycle progression and cell proliferation, which is in accordance to the OTAspecific<br />

increase in cell proliferation and with signs <strong>of</strong> regeneration.<br />

4. Short-term treatment with MAMAc only resulted in a small number <strong>of</strong> deregulated<br />

genes in male Eker and wild-type rats, when compared to AA and OTA treatment,<br />

which was in accordance with the lack <strong>of</strong> effects on pathology or cell proliferation in<br />

male rats.<br />

5. Carcinogen treatment generally resulted in a higher number <strong>of</strong> deregulated genes<br />

in Tsc2 mutant Eker rats compared to wild type rats. However, when compared to<br />

the respective controls, similar pathways were affected. Solely OTA-treated Eker<br />

but not wild type rats demonstrated a number <strong>of</strong> additional genes pointing to an<br />

OTA-mediated ERK-dependent inactivation <strong>of</strong> the Tsc2 tumor suppressor gene.<br />

This finding corresponded well with the significant manifestation <strong>of</strong> preneoplastic<br />

lesions, exclusively seen in OTA-treated Eker rats after 14 days <strong>of</strong> exposure.<br />

From these findings it was concluded that gene expression pr<strong>of</strong>iling allows an in-depth<br />

mechanistic insight into earliest cellular alterations after carcinogen exposure. This notion<br />

was corroborated by the finding that deregulated genes reflect the majority <strong>of</strong> cellular<br />

pathways which are known to be involved in the carcinogenic properties <strong>of</strong> AA and OTA.<br />

Table 9.1 summarizes these pathways, and compares the here presented findings to the<br />

specific results published elsewhere (for further references see Table 9.1). In conclusion,<br />

the findings from chapter 3 and 4 demonstrate that microarrays are sufficient to identify a<br />

compound-specific mode <strong>of</strong> action in the kidney already after just 1-14 days <strong>of</strong> exposure.<br />

169


Chapter 9: Overall Discussion and Future Perspectives<br />

Table 9.1: Summary <strong>of</strong> pathways, suspected to be involved in AA-, OTA- or MAMAc-induced<br />

renal toxicity and carcinogenicity, and <strong>of</strong> genes that may be involved in the suspected<br />

pathways or downstream responses (for details see chapter 3 and 4 and GEO<br />

repository (http://www.ncbi.nlm.nih.gov/projects/geo; accession number: GSE5923).<br />

AA<br />

OTA<br />

Major pathways previously<br />

described in chronic in vivo<br />

or in vitro<br />

Phase I metabolic activation <strong>of</strong><br />

AA (Arlt et al. 2002; Kohara et<br />

al. 2002; Li et al. 2006)<br />

DNA damage (Arlt et al. 2002;<br />

Lord et al. 2004)<br />

Caspase-3 activation<br />

(Balachandran et al. 2005)<br />

Mutations in H-ras and p53<br />

(Schmeiser et al. 1991; Arlt et<br />

al. 2002)<br />

Activation <strong>of</strong> cyclin D1/cdk4<br />

(Chang et al. 2006)<br />

Transcriptional repression <strong>of</strong><br />

Nrf2 target genes (Marin-Kuan<br />

et al. 2006)<br />

Increased histone deacetylase<br />

activity and down regulation <strong>of</strong><br />

RNA synthesis (Luhe et al.<br />

2003; Marin-Kuan et al. 2006)<br />

Transcriptional repression <strong>of</strong><br />

HNF4alpha target genes<br />

(Marin-Kuan et al. 2006)<br />

Activation <strong>of</strong> the mitogenic<br />

ERK1/2 pathway (Horvath et<br />

al. 2002; Marin-Kuan et al.<br />

2007)<br />

Activation <strong>of</strong> the stress-<br />

inducible JNK and p38<br />

pathway (Gekle et al. 2000;<br />

Rached et al. 2006)<br />

Activation <strong>of</strong> the NFkappa B<br />

pathway (Rached et al. 2006)<br />

Impairment <strong>of</strong> cellular Ca 2+<br />

and cAMP homeostasis<br />

(Rahimtula and Chong 1991;<br />

Benesic et al. 2000)<br />

Suggested downstream<br />

effects causally related to<br />

deregulated pathways<br />

Deregulated genes found in this<br />

study, supportive for previous<br />

described pathways and<br />

downstream effects<br />

DNA reactivity Up: AKR1B8, AKR7A3, EPHX1,<br />

CYP4A10, CYP51, NQO1, GNMT,<br />

CES2, CES3<br />

Cell cycle arrest, DNA repair Up: MGMT, PHLDA3, CDKN1A,<br />

SNK, CCNG1, MDM2, FTHFD,<br />

PRODH, OXR1<br />

Down: CDCA3, CCNB1, HMMR,<br />

CDC2, CKS2,<br />

Apoptosis, decreased cell<br />

survival and proliferation<br />

Cell survival and proliferation Not detectable<br />

Cell cycle progression and<br />

proliferation<br />

Impaired cellular defense<br />

against oxidative stress,<br />

enhanced oxidative stress,<br />

DNA-damage<br />

Reduced transcription and<br />

protein expression, gene<br />

silencing<br />

Reduced biotransformation,<br />

decreased detoxification and<br />

excretion<br />

Active mitogenic IGF-MAPK<br />

system, cell survival, cell<br />

transformation<br />

Up: TNFAIP8, HSPA5<br />

Down: USP2, SNN, Ki-67, FRZB,<br />

IGF1, CTGF, TGM2, NAB2,<br />

SFRP2, KLK7<br />

Not detectable<br />

Up: KEAP1, SUPT16H, CHEK2,<br />

MDM2, RBBP6, SEPP1, GGTLA1,<br />

TXNRD1<br />

Down: CN1, HSP40-3, MSRA<br />

Down: H2AA, MYST1, RPP25,<br />

POLD4, NR1D2, KLF13, ID1<br />

Down: CES1B, CES3, AADAC,<br />

CYP2T1, CYP2F4, WBSCR21,<br />

ALDH6A1 , CML1, NAT8, GGT6,<br />

MGST1, GNMT, OCTN1, OAT1,<br />

OCT1<br />

Up: PI3CB, AKT2<br />

Apoptosis Up: JNK2, STAT1, CYBB<br />

Down: CFLAR<br />

Suppressed apoptosis, G1/S<br />

cell cycle transition: Cell<br />

proliferation<br />

Disturbance <strong>of</strong> hormonal Ca 2+<br />

signaling, leading to altered cell<br />

proliferation<br />

Up: GMFB, NFE2L1<br />

Up: CALB, CALR, ITPKB,<br />

PRKAR2A, PPP1R2, CNB, NCX1,<br />

GNAI3, CHP, MPP2<br />

Down: SMP-30<br />

170


MAMAc<br />

Activated TGFß signaling<br />

(Gagliano et al. 2005; Sauvant<br />

et al. 2005)<br />

Spontaneous hydrolysis to the<br />

DNA-reactive<br />

methyldiazonium ion<br />

(Matsumoto and Higa 1966;<br />

Shank and Magee 1967; Sohn<br />

et al. 2001)<br />

Inhibited RNA synthesis<br />

(Zedeck et al. 1970)<br />

Chapter 9: Overall Discussion and Future Perspectives<br />

Altered cell proliferation and<br />

growth control, epithelialmesenchymal-transition,<br />

fibrosis<br />

DNA alkylation, cell cycle arrest,<br />

DNA damage repair, Apoptosis<br />

Up: TGFBR2, SMAD1, SMAD3,<br />

SMAD4, CTNNB1, PDGFC, HAI-1,<br />

NPHS1, TIMP3, DPT, CD44,<br />

NOTCH2<br />

Not detectable<br />

Tumor suppression and others Down: PLAGL1, NR1D1, NR1D2,<br />

NFIX, FKHL18, DBP, BHLHB3,<br />

BAT1A<br />

9.1.2 Prerequisite II: Genotoxic and non-genotoxic renal carcinogens<br />

can be distinguished by early gene expression pr<strong>of</strong>iles<br />

As demonstrated in chapter 3, 1 to 14 days <strong>of</strong> treatment with AA clearly resulted in the upregulation<br />

<strong>of</strong> numerous genes involved in DNA damage response, cell cycle arrest and<br />

apoptosis, indicative for a compound-induced DNA-damaging (genotoxic) action (Branzei<br />

and Foiani 2008) In contrast, gene expression pr<strong>of</strong>iles <strong>of</strong> OTA-treated rats point to an<br />

indirect genotoxic mode <strong>of</strong> action due to increased oxidative stress, as indicated by a broad<br />

down-regulation <strong>of</strong> genes involved in anti-oxidant defense. Compared to the predominant<br />

detection <strong>of</strong> genes involved in DNA damage response in AA-treated rats, OTA treatment<br />

resulted in a marginal deregulation <strong>of</strong> these gene groups, which suggests a secondary role<br />

<strong>of</strong> oxidative DNA damage in OTA-induced kidney cancer. More prominent, OTA treatment<br />

resulted in the additional up-regulation <strong>of</strong> genes involved in mitogenic processes, such as<br />

cell cycle progression, cell survival and proliferation (for details see chapter 3). Already<br />

these findings indicate that gene expression pr<strong>of</strong>iling may allow the prediction <strong>of</strong> genotoxic<br />

or non-genotoxic/ indirect genotoxic carcinogenic potential for a compound in short-term<br />

studies.<br />

This interpretation is further supported by previous reports showing that similar groups <strong>of</strong><br />

genes (indicative either for a genotoxic or and non-genotoxic mode <strong>of</strong> action) were<br />

deregulated in rat liver after short-term exposure to different known genotoxic or nongenotoxic<br />

compounds, respectively (Ellinger-Ziegelbauer et al. 2004; Ellinger-Ziegelbauer<br />

et al. 2005). Specifically, the genotoxic liver carcinogens 2-nitr<strong>of</strong>luorene,<br />

dimethylnitrosamine; 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone and aflatoxin B1 all<br />

resulted in the up-regulation <strong>of</strong> p53 target genes such as CDKN1A, MGMT, CCNG1, or<br />

MDM2. Notably, these genes were also up-regulated in kidneys <strong>of</strong> short-term AA-treated<br />

rats (see chapter 3). In contrast, gene expression pr<strong>of</strong>iles after treatment with non-<br />

171


Chapter 9: Overall Discussion and Future Perspectives<br />

genotoxic liver carcinogens methapyrilene, diethylstilbestrol, Wy-14643 and<br />

piperonylbutoxide appeared more heterogeneous, with a slight emphasis on de-regulated<br />

genes involved in DNA replication and cell cycle progression, cell survival and proliferation<br />

(Ellinger-Ziegelbauer et al. 2005; Ellinger-Ziegelbauer et al. 2008). Consistently, such a<br />

gene expression pr<strong>of</strong>ile was also found in kidneys <strong>of</strong> rats after OTA treatment (Chapter 3).<br />

In contrast to the above findings, 1-14 days <strong>of</strong> treatment with the known genotoxic, DNAalkylating<br />

compound MAMAc did not result in characteristic gene expression pr<strong>of</strong>iles<br />

previously found for other genotoxic, or non-genotoxic compounds. In a predictive<br />

approach, this data would imply a non-carcinogenic action <strong>of</strong> MAMAc, at least at the low<br />

doses used for this study. However, the additional detection <strong>of</strong> an accumulating amount <strong>of</strong><br />

pro-mutagenic O6-methylguanine adducts by LC-MS/MS in the same samples used for<br />

gene expression pr<strong>of</strong>iling corroborates the potentially genotoxic, DNA-alkylating<br />

mechanism <strong>of</strong> MAMAc in Eker rats (see chapter 4). This however would suggest that<br />

microarray analysis might not be sensitive enough to detect the consequences <strong>of</strong> low but<br />

possibly relevant amounts <strong>of</strong> DNA adducts. Another explanation for the lack <strong>of</strong> deregulation<br />

<strong>of</strong> genes involved in DNA damage repair and apoptosis could be that basal expression <strong>of</strong><br />

genes involved in DNA repair and/or apoptosis might be sufficient to deal with low amounts<br />

<strong>of</strong> DNA adducts, at least until a certain threshold dose or duration <strong>of</strong> exposure is exceeded.<br />

Together, the findings presented here suggest that gene expression pr<strong>of</strong>iling in<br />

combination with the analysis <strong>of</strong> cell proliferation and histopathology may allow the<br />

distinction <strong>of</strong> genotoxic from non-genotoxic modes <strong>of</strong> action in short-term studies. However,<br />

microarray analyses may require a minimum amount <strong>of</strong> DNA damage, resulting in a<br />

detectable up-regulation <strong>of</strong> DNA damage response genes. More research on other renal<br />

carcinogens is therefore needed to verify these findings and also to elucidate whether a<br />

carcinogen can be clearly distinguished from nephrotoxic/non-carcinogenic mechanisms in<br />

short term assays.<br />

9.1.3 Prerequisite III: Manifestation <strong>of</strong> early compound-induced<br />

expression changes in kidney tumors.<br />

Eker rats were chronically exposed with AA, OTA, or MAMAc for either 3 or 6 months.<br />

Prolonged exposure should reveal whether early gene expression changes, indicative for a<br />

genotoxic, non-genotoxic/ indirect genotoxic, or non-carcinogenic mode <strong>of</strong> action, would<br />

indeed manifest in the onset or absence <strong>of</strong> renal tumors, respectively. Eker rats were<br />

chosen as the only animal model in these chronic studies out <strong>of</strong> both ethical and financial<br />

172


Chapter 9: Overall Discussion and Future Perspectives<br />

considerations. Specifically, various studies have demonstrated a substantially higher<br />

susceptibility <strong>of</strong> Eker rats towards genotoxic and non-genotoxic carcinogens, resulting in an<br />

earlier manifestation and higher number <strong>of</strong> carcinogen-induced renal pre-neoplastic and<br />

neoplastic lesions compared to wild-type rats (Walker et al. 1992; Wolf et al. 1998; Wolf et<br />

al. 2000). Thus, the duration <strong>of</strong> the study could be significantly shortened and fewer<br />

animals and less compound could be used.<br />

As demonstrated in chapter 4 and 6, prolonged carcinogen exposure corroborated the<br />

findings <strong>of</strong> the short-term assay: Both AA and OTA treatment resulted in a significant<br />

increase in total numbers <strong>of</strong> preneoplastic lesions, and the absence there<strong>of</strong> in MAMActreated<br />

male Eker rats. Moreover, in accordance with previous in vivo studies using AA and<br />

OTA, a similar sex-specific susceptibility towards both carcinogens was found; pointing to a<br />

compound-specific carcinogenic effect <strong>of</strong> AA and OTA in Eker rats (for details see chapter<br />

6 and chapter 8).<br />

A sex-specific susceptibility towards a carcinogenic action <strong>of</strong> MAMAc has not been<br />

described before. The findings <strong>of</strong> this thesis point to a higher susceptibility <strong>of</strong> female rats,<br />

as a significant increase <strong>of</strong> MAMAc-induced preneoplastic lesions was solely observed in<br />

female rats. In male Eker rats, prolonged treatment with MAMAc did not manifest in a<br />

significant increase <strong>of</strong> preneoplastic lesions. This was in accordance with the gene<br />

expression pr<strong>of</strong>iles derived from short-term treated males that did not point to a genotoxic<br />

mode <strong>of</strong> action <strong>of</strong> MAMAc, further corroborating the predictive potential <strong>of</strong> short-term<br />

assays. The lack <strong>of</strong> a tumor-initiating action was surprising since MAMAc has been<br />

identified as a potent genotoxic renal carcinogen in male rats (Laqueur 1964; Laqueur and<br />

Matsumoto 1966; Hirono et al. 1968; Fukunishi et al. 1971). However, all previous studies<br />

on the carcinogenic action <strong>of</strong> MAMAc have used acute high doses (25 to 500 mg/kg bw),<br />

compared to the here shown study using low and chronic doses (250 µg/kg bw). It thus<br />

seems possible that tumor initiation by MAMAc might depend on a threshold dose<br />

ultimately leading to a sufficient amount <strong>of</strong> DNA adducts which then might result in<br />

transforming mutations in critical genes and tumor initiation.<br />

Importantly, although 6 month treatment with MAMAc did not result in an increased number<br />

<strong>of</strong> preneoplastic lesions in males, they showed an increased incidence and higher total<br />

number <strong>of</strong> neoplastic lesions, compared to the respective control rats (for details see<br />

chapter 4). These findings could therefore point to a differential influence <strong>of</strong> MAMAc on<br />

early and later tumor stages, and may be explained by the here detected MAMAc-induced<br />

up-regulation <strong>of</strong> the TGF-ß pathway. This pathway is known to function as a tumor<br />

suppressor at early stages <strong>of</strong> kidney cancer but it can also promote tumor progression and<br />

173


Chapter 9: Overall Discussion and Future Perspectives<br />

metastasis in later stages (Gold 1999; Laping et al. 2007). These findings may therefore<br />

indicate that short-term gene expression pr<strong>of</strong>iling could also be a useful tool to predict the<br />

carcinogenic potential <strong>of</strong> compounds acting on later stages <strong>of</strong> carcinogenesis.<br />

9.1.4 Summary Part I<br />

In summary, the initial hypothesis <strong>of</strong> this thesis, that short-term in vivo assays, using<br />

microarrays as detection system, can be used as a sensitive and specific tool to predict the<br />

renal carcinogenic potential <strong>of</strong> test compounds in the kidney, is corroborated by the here<br />

presented data. However, additional work needs to be done to evaluate gene expression<br />

pr<strong>of</strong>iles in response to different dose-regimens, to also elucidate whether one compound, in<br />

a dose-dependent manner, can simultaneously or successively adopt both non-genotoxic<br />

and genotoxic mechanisms.<br />

Furthermore it needs to be established whether short-term gene expression pr<strong>of</strong>iles would<br />

indeed allow a reliable detection <strong>of</strong> carcinogens that target genes and pathways important<br />

for later stages <strong>of</strong> renal carcinogenesis. This however would require a more detailed<br />

understanding <strong>of</strong> signaling cascades involved in tumor promotion and progression, and the<br />

impact <strong>of</strong> renal carcinogens on these processes. This question was further elucidated in the<br />

second part <strong>of</strong> this thesis and is discussed in detail the following section.<br />

174


Chapter 9: Overall Discussion and Future Perspectives<br />

9.2 Cellular Signaling Cascades in Preneoplastic Lesion<br />

Progression<br />

The two-year rodent bioassay is mainly based on the detection <strong>of</strong> end-stage adenomas and<br />

carcinomas. Shorter in vivo bioassays will however no longer be based on the detection <strong>of</strong><br />

end-stage tumors, but rather have to use specific molecular, predictive endpoints. Clearly,<br />

however, such predictive tests would require a detailed understanding <strong>of</strong> mechanisms<br />

involved in tumor initiation, promotion and progression.<br />

Currently, mechanisms involved in multistage kidney carcinogenesis are only poorly<br />

understood. Thus, the next aim <strong>of</strong> this thesis was to identify and characterize signaling<br />

pathways involved in the progression <strong>of</strong> early preneoplastic renal lesions (for details see<br />

chapter 3 and 6). This should further allow to:<br />

• Analyze the impact <strong>of</strong> genotoxic and non-genotoxic renal carcinogens on these<br />

pathways<br />

• Determine, whether these genes and pathways could be detected in short-term<br />

assays<br />

Since only prolonged treatment with AA and OTA, but not with MAMAc, resulted in a<br />

compound-specific increase <strong>of</strong> total numbers <strong>of</strong> bATs and bAHs in males, gene expression<br />

studies from microdissected lesions were restricted to male AA- and OTA-treated rats and<br />

vehicle controls. For technical reasons, later tumor stages (adenomas and carcinomas)<br />

were not further addressed, since the cryo-conserved tissue designated for laser<br />

microdissection did not comprise enough replicate tumors in each dose group for proper<br />

statistical analysis.<br />

First, a protocol was established that allowed a reproducible and reliable analysis <strong>of</strong> gene<br />

expression pr<strong>of</strong>iles from laser-microdissected preneoplastic lesions (for details see chapter<br />

5). Using this protocol, gene expression pr<strong>of</strong>iles <strong>of</strong> different stages <strong>of</strong> preneoplastic lesions<br />

(bATs and bAHs) and healthy tubules <strong>of</strong> AA-, OTA- and vehicle-treated male Eker rats<br />

were analyzed and revealed the following key findings:<br />

1. Consistent with the initial hypothesis, the here presented findings demonstrate for<br />

the first time that large-scale gene expression pr<strong>of</strong>iling from microdissected earliest<br />

stages <strong>of</strong> renal preneoplastic lesions is possible (for details see chapter 5 and 6).<br />

175


Chapter 9: Overall Discussion and Future Perspectives<br />

2. As expected at the beginning <strong>of</strong> this thesis, preneoplastic lesions showed a<br />

massive deregulation <strong>of</strong> genes compared to healthy tissue <strong>of</strong> control rats, clearly<br />

demonstrating that tumor cells are different from their cellular origin (chapter 6).<br />

3. In contrast to the initial hypothesis, different stages <strong>of</strong> microdissected preneoplastic<br />

lesions (bATs and bAHs) were similar in their transcriptome chapter 6).<br />

4. Unexpectedly, comparisons <strong>of</strong> gene expression pr<strong>of</strong>iles from microdissected<br />

preneoplastic lesions <strong>of</strong> vehicle-treated with AA- and OTA-treated rats revealed<br />

only marginal differences. Thus, carcinogen treatment does not seem to influence<br />

gene deregulation once a preneoplastic lesion has manifested.<br />

5. Chronic treatment with AA or OTA did not substantially change gene expression in<br />

microdissected healthy tubules, which was in contrast to the overt carcinogeninduced<br />

gene expression changes in short-term treated Eker rats (see chapter 3<br />

and 6).<br />

The above findings are discussed in detail in the respective manuscripts. The next chapter<br />

will summarize all findings and propose a hypothetical model <strong>of</strong> carcinogen-induced renal<br />

tumorigenesis in Eker rats and potentially also wild-type strains. This model delineates the<br />

pivotal roles <strong>of</strong> carcinogen-induced tubular damage, regeneration and adaptation<br />

processes for the development <strong>of</strong> kidney cancer (see chapter 9.2.1), and proposes a<br />

predictive potential <strong>of</strong> preneoplastic lesions for the carcinogenic outcome <strong>of</strong> the rodent<br />

bioassays (see chapter 9.2.2).<br />

9.2.1 Pivotal role <strong>of</strong> carcinogen-induced tubular damage, regeneration<br />

and adaptation processes for the development <strong>of</strong> kidney cancer<br />

Based on the differing gene expression pr<strong>of</strong>iles from kidney homogenates and healthy<br />

tubules <strong>of</strong> short-term vs. chronically treated rats, respectively, a hypothetical model <strong>of</strong><br />

carcinogen-induced renal tumorigenesis is proposed (Figure 9.1, A-E) This model assumes<br />

that proximal tubules <strong>of</strong> a similar microscopic phenotype generally differ in their sensitivity<br />

towards renal carcinogens (Figure 9.1, A). Such differences in carcinogen sensitivity could<br />

be based on variations in the cellular environment (e.g. amount <strong>of</strong> blood and carcinogen<br />

supply), cellular polymorphisms in carcinogen uptake, metabolic activation, detoxification,<br />

DNA damage repair or many other variations in intracellular carcinogen binding sites.<br />

Depending on their intrinsic sensitivity, some tubules would remain rather unaffected by<br />

carcinogen treatment, while others are sensitive towards genotoxic, non-genotoxic or<br />

176


Chapter 9: Overall Discussion and Future Perspectives<br />

cytotoxic effects. Gene expression pr<strong>of</strong>iling from short-term treated rats will therefore reveal<br />

compound-specific deregulations in sensitive tubules, as shown in chapter 3 and 4).<br />

As demonstrated for AA and OTA, prolonged carcinogen treatment can result in<br />

compound-specific cytotoxic or genotoxic effects accompanied with apoptosis or necrosis<br />

and regenerative cell proliferation (Chapter 6). Regeneration might be based on the<br />

replication <strong>of</strong> less affected or more insensitive neighboring cells (Figure 9.1, B). Thus,<br />

recurring cycles <strong>of</strong> damage and regeneration might also constitute a selection towards<br />

carcinogen-insensitive cells with a healthy phenotype (Figure 9.1, C). Gene expression<br />

pr<strong>of</strong>iles from microdissected healthy tubules <strong>of</strong> carcinogen-treated rats, when compared<br />

with healthy tubules <strong>of</strong> control rats, would thus not demonstrate overt compound-specific<br />

effects even though all healthy tubules are still systemically supplied with carcinogens<br />

(Chapter 6, Figure 6.2). However, a qualitative comparison <strong>of</strong> short-term gene expression<br />

pr<strong>of</strong>iles from kidney homogenates <strong>of</strong> AA- and OTA-treated rats with those derived from<br />

microdissected healthy tubules <strong>of</strong> AA- and OTA-treated rats, respectively, demonstrated<br />

several similarly deregulated genes (Figure 9.2). This finding may point to a low number <strong>of</strong><br />

sensitive healthy tubules remaining in AA- and OTA-treated rats.<br />

Figure 9.1: Overview <strong>of</strong> the hypothetical model <strong>of</strong> chemically-induced renal carcinogenesis in<br />

Eker rats<br />

177


Chapter 9: Overall Discussion and Future Perspectives<br />

Recurring cycles <strong>of</strong> compound-induced cell death and regeneration also allow the fixation<br />

<strong>of</strong> carcinogen-induced or spontaneous mutation and, if critical genes were affected, their<br />

clonal expansion to an atypical tubule (Figure 9.1, D). Thus, the compound-specific amount<br />

<strong>of</strong> damage and regeneration could be an important driving force for the development <strong>of</strong><br />

preneoplastic lesions that determines their total numbers. A correlation between tubular<br />

damage, increased cell proliferation and the total numbers <strong>of</strong> preneoplastic lesions was<br />

demonstrated in chapter 4 and 6 and is further supported by the findings <strong>of</strong> Cunningham<br />

and colleagues (Cunningham et al. 1993), who demonstrated that mutagenic compounds<br />

that increased renal cell proliferation also increased renal tumors, whereas mutagens<br />

having no effect on renal cell proliferation did not.<br />

The carcinogen-induced transformation <strong>of</strong> tubular cells together with the finding that<br />

atypical tubules are insensitive towards carcinogen treatment points to a carcinogen<br />

resistance <strong>of</strong> clonal expanding cells (Figure 9.1, E). A similar phenomenon is already<br />

known as multidrug resistance, where e.g. cancer cells have the ability to become resistant<br />

to chemotherapeutics. This includes protective mechanisms, e.g. an increased efflux <strong>of</strong><br />

drugs or enzymatic deactivation, but could also be the result <strong>of</strong> decreased uptake or<br />

membrane permeability, or <strong>of</strong> altered drug binding sites (Szakacs et al. 2006). Indeed, as<br />

demonstrated in chapter 6, the functional analysis <strong>of</strong> gene expression pr<strong>of</strong>iles derived from<br />

preneoplastic lesions demonstrates a broad down-regulation <strong>of</strong> transporters and metabolic<br />

enzymes previously demonstrated to be involved in AA- and OTA-mediated toxicity and<br />

carcinogenicity (Gekle and Silbernagl 1994; Stiborova et al. 2003; O'Brien and Dietrich<br />

2005), for details see GEO repository, http://www.ncbi.nlm.nih.gov/projects/geo; accession<br />

number: GSE 10608. However, it remains elusive whether the here detected downregulation<br />

<strong>of</strong> genes is a specific feature <strong>of</strong> atypical renal tubular cells, or results from an<br />

unspecific dedifferentiation <strong>of</strong> fast proliferating cells. An additional explanation for the<br />

general insensitivity <strong>of</strong> preneoplastic lesions towards AA- and OTA-treatment could be that<br />

a temporarily reduced blood supply in fast growing pre-neoplastic foci could reduce the<br />

amount <strong>of</strong> carcinogen reaching the cells, at least until angiogenesis starts.<br />

178


Chapter 9: Overall Discussion and Future Perspectives<br />

Figure 9.2: Heatmap <strong>of</strong> compound-specific unions <strong>of</strong> genes found significantly deregulated (red:<br />

up-regulated; green: down-regulated) after carcinogen treatment. Gene expression<br />

pr<strong>of</strong>iles are derived from AA- or OTA-treated Eker or wild type rats after 1 or 14 days<br />

<strong>of</strong> treatment (WT-d1 to EK/d14), and from microdissected healthy tubules <strong>of</strong> 6 month<br />

treated rats (HT). All genes were compared to their corresponding vehicle controls<br />

(n=3 per time-point). The color scale at the left depicts gene expression ratios. Note<br />

that only a qualitative comparison <strong>of</strong> genes is possible, since arrays were not<br />

processed at same time points and only 1mg/kg bw instead <strong>of</strong> 10mg/kg AA were used<br />

for chronic experiments.<br />

The here presented findings suggest that the increase <strong>of</strong> preneoplastic lesions primarily<br />

depends on the initial DNA damaging or nephrotoxic action <strong>of</strong> AA and OTA, while lesion<br />

progression is the result <strong>of</strong> compound-independent clonal expansion (Chapter 3 and 6).<br />

The extent <strong>of</strong> damage and regeneration may also determine the selection rate <strong>of</strong><br />

compound-insensitive cells. This suggests the existence <strong>of</strong> a specific time window where<br />

healthy tubules are still sensitive towards carcinogen treatment and not yet adapted. Such<br />

a critical period <strong>of</strong> carcinogen exposure would imply that extended compound exposure<br />

might add little to the formation <strong>of</strong> additional preneoplastic lesions. Instead, the number <strong>of</strong><br />

preneoplastic lesions formed during this critical period can determine the incidence and<br />

number <strong>of</strong> veritable tumors at later stages.<br />

However, not all carcinogen-induced preneoplastic lesions will progress to neoplastic<br />

lesions. Instead, some preneoplastic lesions may also be self-limiting or even reversible<br />

(Alden and Kanerva 1982; Eustis 1989; Dietrich and Swenberg 1991). The identification <strong>of</strong><br />

179


Chapter 9: Overall Discussion and Future Perspectives<br />

pathways that distinguish self-limiting or reversible preneoplastic lesions from lesions with<br />

true neoplastic potential would therefore greatly improve the predictive value <strong>of</strong><br />

preneoplastic lesions for carcinogenicity testing. Potential genes and pathways are<br />

discussed in the following section.<br />

9.2.2 Predictive potential <strong>of</strong> preneoplastic lesions as tumor precursors<br />

Tumor progression has been suggested to depend on acquiring six essential "hallmarks <strong>of</strong><br />

cancer”, that collectively dictate malignant growth (Hanahan and Weinberg 2000) (Chapter<br />

1.1.3 and Table 9.2). The basic concept <strong>of</strong> Hanahan and Weinberg is well accepted and<br />

may be shared by the majority, if not by all tumor types. However, to date it is not clear<br />

whether some or all <strong>of</strong> these acquired <strong>characteristics</strong> are detectable in preneoplastic<br />

lesions.<br />

Gene expression pr<strong>of</strong>iles derived from different progression stages <strong>of</strong> renal tumors, i.e.<br />

from early basophilic atypical tubules (bAT) were highly similar to later basophilic atypical<br />

hyperplasias (bAHs) (Chapter 6). This raised the hypothesis that early atypical tubules may<br />

already have all prerequisites to develop into neoplastic lesions. Indeed, functional analysis<br />

<strong>of</strong> gene expression pr<strong>of</strong>iles from microdissected renal bATs and bAHs identified groups <strong>of</strong><br />

deregulated genes and pathways that were indicative for all six hallmarks <strong>of</strong> cancer. Table<br />

9.2 summarizes some <strong>of</strong> the functional groups <strong>of</strong> genes and pathways that may give rise to<br />

the respective physiological changes, as proposed by Hanahan and Weinberg. Together,<br />

these findings suggest that the earliest microscopically detectable preneoplastic lesions<br />

from Eker rats may have the potential to develop into a malignant phenotype.<br />

Functional analysis <strong>of</strong> deregulated genes as well as immunohistochemical staining <strong>of</strong><br />

pS6RP, pAKT and FOXO1 in bATs and bAHs <strong>of</strong> control, AA- and OTA-treated Eker rats<br />

further demonstrated an activation <strong>of</strong> the mTOR pathway when compared to healthy tissue<br />

<strong>of</strong> control animals (for details see chapter 6). The latter may result from a functional loss <strong>of</strong><br />

the TSC2 tumor suppressor gene in preneoplastic lesions by genetic or epigenetic events,<br />

leading to a permanent activation <strong>of</strong> the TSC2-mTOR pathway.<br />

180


Chapter 9: Overall Discussion and Future Perspectives<br />

Table 9.2: Genes and pathways linked to the six hallmarks <strong>of</strong> cancer, as suggested by Hanahan<br />

and Weinberg (Hanahan and Weinberg 2000). For specific information see chapter 6<br />

and GEO repository (http://www.ncbi.nlm.nih.gov/projects/geo; accession number:<br />

GSE 10608)<br />

Hallmark <strong>of</strong> cancer Suggested functional gene groups indicative<br />

for the respective hallmarks<br />

Self-sufficiency in<br />

growth signals<br />

Insensitivity to<br />

growth inhibitory<br />

signals<br />

Evasion <strong>of</strong><br />

apoptosis<br />

Limitless replicative<br />

potential<br />

Sustained<br />

angiogenesis<br />

Tissue invasion and<br />

metastasis<br />

Up-regulation <strong>of</strong> growth factors and their<br />

receptors<br />

Significantly deregulated genes, found<br />

in preneoplastic lesions <strong>of</strong> this study,<br />

supportive for the respective hallmark<br />

Up: CTGF, HDGFRP3, IGFBP1, CNTF<br />

Up-regulation <strong>of</strong> oncogenes Up: MYC, NIBAN, ELK3, CASC4<br />

Up-regulation <strong>of</strong> members <strong>of</strong> the SOS-RAS-RAF-<br />

MAPK pathway<br />

Down-regulation <strong>of</strong> tumor suppressor genes or<br />

survival factors<br />

Down-regulation <strong>of</strong> terminal differentiation factors Down: HNF-4A<br />

Up-regulation <strong>of</strong> anti-apoptotic genes and survival<br />

factors<br />

Down-regulation <strong>of</strong> pro-apoptotic genes Down: DNASE1<br />

Up-regulation <strong>of</strong> genes involved in cell cycle<br />

progression<br />

Up: RAB2, RAB3D, RAB31, RAB6, NXN<br />

Down: PINK1, CRYL1, FOXO1A, CHN2<br />

Up: PEA-1, BNIP2, TMBIM1, ARC,<br />

SH3BGRL3, CLU, GAS6, NAP1L1, KITL,<br />

NRTN, EMP1<br />

Up:, CCNB1, CCNB2, CDC2, CDCA3,<br />

CKS2, PCTK2, AURKB, KIF15, KIF22,<br />

PLK4, UBE2C, RMBS2, POLD1, MCM2,<br />

MCM3, PAF<br />

Up-regulation <strong>of</strong> endothelial growth factors Up: PAI1 CXCR4, GP38, DDL4, DSCR1,<br />

Up-regulation <strong>of</strong> smooth muscle components Up: CRP1, PSEL,<br />

Up-regulation <strong>of</strong> proteases or cell contact<br />

inhibition<br />

Up-regulation <strong>of</strong> genes involved in epithelial-<br />

mesenchymal transition (EMT)<br />

Up: tPA, BMP1, CTSK, CAPN2, ST14,<br />

MEP1A, DPP7, KLK12, FXYD5, CD44<br />

Up: VIM, CTGF, ACVR1, TGFBI, TGFB2,<br />

Down-regulation <strong>of</strong> protease inhibitors Down: SERPINF2, HRG<br />

Down-regulation <strong>of</strong> cell adhesion molecules Down: CXN-26, PCLKC, TFF3, TNS<br />

An in-depth description <strong>of</strong> this pathway is given in the introduction <strong>of</strong> chapter 6 and is<br />

summarized in Figure 9.4. Briefly, the mammalian target <strong>of</strong> rapamycin (mTOR) is an<br />

intracellular Ser/Thr kinase that is part <strong>of</strong> two distinct protein complexes called TORC1 and<br />

TORC2. The raptor-containing mTOR complex TORC1 is inhibited by the TSC1/ TSC2<br />

complex, remains rapamycin-sensitive, and mainly activates ribosomal biogenesis,<br />

translation and nutrient import. TORC2 is rapamycin-insensitive and is required for the<br />

organization <strong>of</strong> the cytoskeleton (Mak and Yeung 2004; Jozwiak 2006; Wullschleger et al.<br />

2006). Both TORC1 and TORC2 are key regulators <strong>of</strong> numerous important cellular<br />

processes like cell growth, -survival, -proliferation, -motility and the nutrient-hormonal<br />

signaling network. However, upon loss <strong>of</strong> the tumor suppressor TSC2, e.g. by carcinogeninduced<br />

mutations, mTORC1 activity is aberrantly increased, leading to pathologies such<br />

as cancer, metabolic disorders and cardiovascular diseases (Thomas 2006).<br />

181


Chapter 9: Overall Discussion and Future Perspectives<br />

Under physiological conditions, TSC2 can be phosphorylated and inactivated by AKT in<br />

response to growth factors (e.g. insulin), thus resulting in activation <strong>of</strong> mTORC1. TORC2 in<br />

turn was demonstrated to directly phosphorylate AKT at Ser 473, resulting in the activation<br />

<strong>of</strong> several down-stream pathways, including an indirect activation <strong>of</strong> TORC1 (Wullschleger<br />

et al. 2006). Recent studies added even further complexity by suggesting the existence <strong>of</strong> a<br />

negative feedback loop from the activated TSC2-TORC1-S6K1 pathway to the upstream<br />

insulin-responsive IRS-PI3K-PDK1-AKT pathway (Figure 9.4) (Harrington et al. 2004; Um<br />

et al. 2004). It was previously demonstrated that the mTORC1-dependent negative<br />

feedback loop is still present in benign and growth-limited hamartomas (Kwiatkowski 2003;<br />

Manning et al. 2005). This was in contrast to the findings <strong>of</strong> this study, which point to a<br />

concomitant activation <strong>of</strong> TORC1 and TORC2 signaling and therefore to a disturbance <strong>of</strong><br />

this feedback loop in preneoplastic lesions from Eker rats (for details see chapter 6). In this<br />

scenario, AKT activation due to TORC2 dependent Ser473 phosphorylation (Bellacosa et<br />

al. 2005) would not only further ameliorate the AKT-TSC2-TORC1-dependent oxygen and<br />

energy supply <strong>of</strong> proliferating cells via increased angiogenesis and glycolysis, but would<br />

also reduce AKT-dependent apoptosis and cell cycle progression.<br />

Together with the functional gene expression analysis, pointing to the establishment <strong>of</strong> all<br />

six hallmarks <strong>of</strong> cancer, it might be possible that the simultaneous activation <strong>of</strong> TORC1 and<br />

TORC2 may act as a neoplastic switch in renal preneoplastic lesions. Thus, disturbance <strong>of</strong><br />

the negative feedback loop <strong>of</strong> the AKT-TSC2-mTOR pathway may be indicative <strong>of</strong> a<br />

malignant outcome. This is consistent with previous findings showing that the suppression<br />

<strong>of</strong> phosphatase and tensin homolog PTEN in TSC2 mutant cells, which efficiently blocks<br />

the phosphorylation <strong>of</strong> AKT, reactivates the PI3K-AKT pathway to generate more<br />

aggressive tumors (Ma et al. 2005b). However, further research is needed to verify a direct<br />

mechanistic correlation <strong>of</strong> these findings, for instance by using specific TORC1 or TORC2<br />

inhibitors and a combination there<strong>of</strong> in in vivo carcinogenicity studies. A specific inhibitor <strong>of</strong><br />

TORC1, rapamycin, is already used under the trade name ORISEL® (temsirolimus; Wyeth<br />

USA) to treat patients with advanced RCC. However, and in accordance with the here<br />

presented findings, new evidence suggests that mTOR might have a more central<br />

rapamycin-insensitive role in the pathology <strong>of</strong> some cancers e.g. by hyperactive TORC2<br />

signalling. In Spring 2008, a small molecule dual TORC1/TORC2 kinase inhibitor has<br />

reached clinical trials (OSI-027, OSI Pharmaceuticals, Inc), which might help to further<br />

clarify the role <strong>of</strong> this pathway in tumorigenesis.<br />

182


Chapter 9: Overall Discussion and Future Perspectives<br />

Figure 9.3: Overview <strong>of</strong> the mTOR pathway and its hypothesized function within a proximal<br />

tubule. Modified from (Wullschleger 2006, Thomas, 2006, Mak, 2004, Jacinto, 2004,<br />

Hannah, 2008).<br />

9.2.3 Summary Part II<br />

For the first time, the findings from Eker rats have demonstrated a possible adaptation <strong>of</strong><br />

(damaged and regenerating) healthy tubules and an insensitivity <strong>of</strong> preneoplastic lesions<br />

towards prolonged carcinogen treatment. Furthermore, these data indicate that the<br />

incidence and number <strong>of</strong> veritable tumors is primarily driven by the number <strong>of</strong> preneoplastic<br />

lesions initially formed during a critical period <strong>of</strong> carcinogen exposure. Clonal expansion <strong>of</strong><br />

preneoplastic lesions however seems to be a compound-independent process, and may be<br />

essentially driven by the mTOR pathway. The finding that both, TORC1 and TORC2 are<br />

activated in preneoplastic lesions <strong>of</strong> Eker rats points to a disturbed negative feedback loop<br />

<strong>of</strong> the PI3K-AKT-TSC2-mTOR pathway. Together with the establishment <strong>of</strong> the 6 hallmarks<br />

<strong>of</strong> (malignant) cancer, this finding suggests that the earliest detectable preneoplastic<br />

lesions <strong>of</strong> Eker rats have the potential to overcome growth- and proliferation-limiting<br />

barriers, and thus develop into full neoplasms. <strong>Molecular</strong> <strong>characteristics</strong> <strong>of</strong> the earliest<br />

183


Chapter 9: Overall Discussion and Future Perspectives<br />

detectable preneoplastic lesions might therefore already be indicative for their later<br />

outcome. In conclusion, the use <strong>of</strong> preneoplastic lesions as predictive endpoints in<br />

shortened rodent bioassays seems principally possible.<br />

9.3 Obesity as biasing factor for a reliable human risk<br />

assessment from rodent bioassays<br />

A large number <strong>of</strong> previous reports, as well as findings presented within this thesis, have<br />

demonstrated an important role <strong>of</strong> the activated mTOR pathway in certain types <strong>of</strong> rodent<br />

and human cancer (see 9.2.2). Next to its role in tumorigenesis, the mTOR pathway is also<br />

known to be a central regulator for the nutrient-hormonal signaling network and activation<br />

<strong>of</strong> the TORC1 complex is commonly detected in in vivo and in vitro models <strong>of</strong> obesity<br />

(Castaneda et al. 2008; Cota et al. 2008; Gulati et al. 2008. Thus, it was hypothesized that<br />

hyperactivation <strong>of</strong> the mTOR pathway might be a missing link between obesity and kidney<br />

cancer (Thomas, 2006). However, the exact impact <strong>of</strong> chronic high fat diet exposure on<br />

kidney cancer and the renal mTOR pathway remains widely elusive.<br />

In the final part <strong>of</strong> this thesis, it was therefore hypothesized that prolonged high fat diet<br />

exposure leads to the manifestation <strong>of</strong> severe renal pathology with early markers <strong>of</strong> renal<br />

carcinogenesis. Specifically, this thesis aimed to dissect the impact <strong>of</strong> high body adiposity<br />

with its known co-morbidities and disease consequences versus the direct impact <strong>of</strong> dietary<br />

lipids on cellular renal pathways involved in renal carcinogenesis, such as the mTOR-S6K<br />

pathway. Renal pathology in a diet-sensitive subpopulation ((DIOsens) characterized by a<br />

morbidly increased body weight) was examined and compared to a diet-resistant<br />

subpopulation ((DIOres) with moderately increased body weight) <strong>of</strong> male Wistar rats as well<br />

as to chow fed control rats with normal body weight. This resulted in the following key<br />

findings (for details see chapter 7).<br />

1. DIOsens and DIOres rats but not control rats demonstrated marked tubular<br />

damage, regenerative cell proliferation and chronic progressive nephropathy<br />

(CPN). CPN was not only associated with a simple tubular hyperplasia, but also<br />

with preneoplastic lesions, which might have the potential to manifest into solid<br />

tumors at later stages <strong>of</strong> life.<br />

2. In both DIOsens and DIOres rats positive staining <strong>of</strong> phosphorylated ribosomal<br />

protein S6 (P-RPS6), a downstream effecter <strong>of</strong> activated TORC1, was detected in<br />

184


Chapter 9: Overall Discussion and Future Perspectives<br />

nearly all cells <strong>of</strong> regenerating tubules within the CPN lesions, and especially in<br />

tubules with an atypical phenotype.<br />

3. A similar degree <strong>of</strong> histopathological changes in DIOsens and DIOres rats<br />

suggested a more prominent role <strong>of</strong> the dietary fat content, rather than the degree<br />

<strong>of</strong> adiposity, for the etiology <strong>of</strong> renal pathology and TORC1 activation.<br />

Together these findings demonstrate a clear adverse effect <strong>of</strong> dietary lipids on renal<br />

morphology and further implicate that dietary lipids may act via similar mechanism like<br />

nephrotoxic compounds. The findings <strong>of</strong> chapter 3, 4 and 6 implicate that renal tissue<br />

damage, inflammation and regeneration are important factors contributing to the onset and<br />

progression <strong>of</strong> RCC. High-fat diet-induced renal damage therefore may further propagate<br />

the fixation <strong>of</strong> spontaneous mutations and progression <strong>of</strong> pre-neoplastic lesions into solid<br />

renal tumors. Notably, preneoplastic lesions are frequently found within the CPN lesions<br />

and might be indicative for a cancer promoting action <strong>of</strong> dietary lipids. However, to date it<br />

cannot be determined whether such preneoplastic lesions would further develop into gross<br />

tumors. Instead, the observed lesions could be self-limiting or even reversible, since no<br />

adenomas or carcinomas could be detected in this study (see chapter 9.2). Nevertheless,<br />

the lack <strong>of</strong> gross tumors was not surprising and indeed was not expected after 11 month <strong>of</strong><br />

high-fat diet exposure, since full adenomas and carcinomas in rodent bioassays are <strong>of</strong>ten<br />

only detectable after 15 to 18 months <strong>of</strong> carcinogen treatment (Haseman et al. 2001).<br />

The findings <strong>of</strong> chapter 7 have demonstrated an increased amount <strong>of</strong> phosphorylated<br />

RPS6 in atypical and regenerating tubules <strong>of</strong> DIOsens and DIOres rats, but not in<br />

unaffected tissue or in control animals. These preliminary findings may point to a role <strong>of</strong><br />

TORC1 in dietary lipid-induced renal damage and regeneration. However, it cannot be<br />

concluded whether high fat diet directly results in an activation <strong>of</strong> TORC1 and RPS6<br />

phosphorylation in renal cells (e.g. via pathways summarized in Figure 9.4). Alternatively,<br />

TORC1 activation could simply be a secondary effect <strong>of</strong> cell regeneration, e.g. via<br />

stimulation with extracellular growth factors like IGF-1 (Moore et al. 2008). Thus, clearly<br />

more research is needed to clarify whether the nephrotoxic action <strong>of</strong> high fat diet may be<br />

involved in the onset or progression <strong>of</strong> spontaneous mutations. This question could be<br />

answered in an in vivo tumor initiation-promotion study, using e.g. aristolochic acid as<br />

kidney specific tumor initiator and high fat diet as possible promoter. In addition, further<br />

studies should be conducted in vitro in primary kidney cells or established kidney cell lines,<br />

to clarify whether direct exposure to various fatty acids might directly stimulate renal<br />

TORC1 or TORC2.<br />

185


Chapter 9: Overall Discussion and Future Perspectives<br />

Considering that obesity has reached epidemic proportions, further elucidating the impact<br />

<strong>of</strong> high fat diet on renal and other types <strong>of</strong> cancer seems not only important under a<br />

medical aspect, but also regarding carcinogenicity testing in rodents. The choice <strong>of</strong> diet can<br />

severely affect the results <strong>of</strong> the rodent bioassay. For instance ad libitum-feeding <strong>of</strong> a high<br />

caloric diet considerably enhanced weight gain, increased background tumor development<br />

and non-neoplastic lesions, and decreased the life-span <strong>of</strong> rodents, compared to those fed<br />

with a caloric restricted chow (Keenan et al. 1995; Keenan et al. 1996). Thus, the dosage<br />

and composition <strong>of</strong> a diet can significantly influence the effects <strong>of</strong> a test compound,<br />

possibly by enhancing tubular damage and regeneration or by (synergistic) a”ctivation <strong>of</strong><br />

the mTOR pathway.<br />

In conclusion, high-fat diet exposure with high body adiposity clearly represents principal<br />

biasing factors for a reliable human risk assessment <strong>of</strong> compounds in rodent bioassays. It<br />

seems entirely possible that previous carcinogenicity testing in rodents underestimated the<br />

potential impact <strong>of</strong> a carcinogen in human subpopulations exposed to a high fat/ high<br />

calorie environment. Future studies should address this important issue, and should clarify<br />

whether carcinogen exposure in obese human subpopulations indeed could be linked to an<br />

enhanced number and incidence <strong>of</strong> renal cancer.<br />

9.4 Synopsis<br />

Evaluating the carcinogenic potential <strong>of</strong> test compounds by life-time rodent bioassays is<br />

time-consuming and associated with high financial costs, high numbers <strong>of</strong> animals, and a<br />

high amount <strong>of</strong> compound is required for continuous dosing over the entire period <strong>of</strong> the<br />

assay. With respect to the kidney, the findings <strong>of</strong> this thesis suggest that it could be<br />

possible to shorten rodent bioassays and to increase the specificity <strong>of</strong> these assays by<br />

using predictive biomarkers in short-term, or intermediate in vivo assays. The key findings<br />

<strong>of</strong> this study, (a) that microarrays allow the detection and distinction <strong>of</strong> genotoxic and nongenotoxic<br />

carcinogens in short-term assays and (b) that the incidence and number <strong>of</strong><br />

veritable tumors is primarily driven by the number <strong>of</strong> preneoplastic lesions initially formed<br />

during a critical period <strong>of</strong> exposure. If the above hypothesis would be transferable to other<br />

organs, then the following possibilities seem likely:<br />

• Predictive biomarkers could serve as early and specific indicators for a genotoxic or<br />

non-genotoxic carcinogenic action <strong>of</strong> test compounds in short-term (prescreening)<br />

186


Chapter 9: Overall Discussion and Future Perspectives<br />

assays. With short term assays the use <strong>of</strong> carcinogenic compounds could be<br />

reduced.<br />

• The continuous dosage <strong>of</strong> animals could be limited to the “critical” time window<br />

where cells are not yet adapted to the carcinogen exposure. Thus, much shorter<br />

time-period <strong>of</strong> exposure with a concomitant reduction <strong>of</strong> pain and distress for the<br />

animals could be achieved.<br />

• Predictive biomarkers might identify the carcinogenic potential <strong>of</strong> preneoplastic<br />

lesions by distinguishing between a potential malignant or self-limiting phenotype.<br />

Furthermore, specific markers may help to distinguish carcinogen-induced<br />

preneoplastic lesions from spontaneous background lesions. Such mechanistic<br />

biomarkers involved in rodent and human preneoplastic lesion progression (e.g. the<br />

mTOR pathway) could therefore serve as improved predictors for the neoplastic<br />

outcome in rats and the potential risk in humans.<br />

• By further specifying the carcinogenic potential <strong>of</strong> preneoplastic lesions, these could<br />

indeed serve as reliable endpoints for carcinogenicity testing and thus shorten the<br />

assay. In addition, reduced mortality in shorter assays would also allow a reduction in<br />

the number <strong>of</strong> animals. Moreover, residual animals would not suffer from extensive<br />

tumor growth.<br />

In general, higher efficiencies <strong>of</strong> the current safety testing paradigm (shorter assays with<br />

fewer animals) would lead to the reduction <strong>of</strong> financial costs for product development. In<br />

conclusion, the here presented findings suggest that the standard rodent bioassay can<br />

indeed be improved, mainly by shortening the duration <strong>of</strong> the life-time rodent bioassay, and<br />

by using novel methodology such as gene array. Ultimately, this thesis provides<br />

encouraging first results that may one day help to establish a new rodent bioassay that<br />

ultimately reduces and refine refines and even replaces animal experiments.<br />

187


Chapter 10: References<br />

Abdollahi A (2007) LOT1 (ZAC1/PLAGL1) and its family members: mechanisms and functions. J Cell<br />

Physiol 210:16-25<br />

Aguila MB, Mandarim-De-Lacerda CA (2003) Effects <strong>of</strong> chronic high fat diets on renal function and cortical<br />

structure in rats. Exp Toxicol Pathol 55:187-195<br />

Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF (2003) Prospective identification <strong>of</strong><br />

tumorigenic breast cancer cells. Proc Natl Acad Sci U S A 100:3983-3988<br />

Albanell J, Dalmases A, Rovira A, Rojo F (2007) mTOR signalling in human cancer. Clin Transl Oncol<br />

9:484-493<br />

Alden CL, Kanerva RL (1982) Reversibility <strong>of</strong> renal cortical lesions induced in rats by high doses <strong>of</strong><br />

nitrilotriacetate in chronic feeding studies. Food Chem Toxicol 20:935-937<br />

Altunkaynak ME, Ozbek E, Altunkaynak BZ, Can I, Unal D, Unal B (2008) The effects <strong>of</strong> high-fat diet on<br />

the renal structure and morphometric parametric <strong>of</strong> kidneys in rats. J Anat 212:845-852<br />

Ames BN, Gold LS (1990) Chemical carcinogenesis: too many rodent carcinogens. Proc Natl Acad Sci U<br />

S A 87:7772-7776<br />

Arlt VM, Pfohl-Leszkowicz A, Cosyns J, Schmeiser HH (2001) Analyses <strong>of</strong> DNA adducts formed by<br />

ochratoxin A and aristolochic acid in patients with Chinese herbs nephropathy. Mutat Res<br />

494:143-150<br />

Arlt VM, Stiborova M, Schmeiser HH (2002) Aristolochic acid as a probable human cancer hazard in<br />

herbal remedies: a review. Mutagenesis 17:265-277<br />

Arlt VM, Stiborova M, vom Brocke J, Simoes ML, Lord GM, Nortier JL, Hollstein M, Phillips DH, Schmeiser<br />

HH (2007) Aristolochic acid mutagenesis: molecular clues to the aetiology <strong>of</strong> Balkan endemic<br />

nephropathy-associated urothelial cancer. Carcinogenesis 28:2253-2261<br />

Armitage JA, Lakasing L, Taylor PD, Balachandran AA, Jensen RI, Dekou V, Ashton N, Nyengaard JR,<br />

Poston L (2005) Developmental programming <strong>of</strong> aortic and renal structure in <strong>of</strong>fspring <strong>of</strong> rats fed<br />

fat-rich diets in pregnancy. J Physiol 565:171-184<br />

Armitage P, Doll R (1954) The age distribution <strong>of</strong> cancer and a multi-stage theory <strong>of</strong> carcinogenesis. Br J<br />

<strong>Cancer</strong> 8:1-12<br />

Attoub S, Noe V, Pirola L, Bruyneel E, Chastre E, Mareel M, Wymann MP, Gespach C (2000) Leptin<br />

promotes invasiveness <strong>of</strong> kidney and colonic epithelial cells via phosphoinositide 3-kinase-, rho-,<br />

and rac-dependent signaling pathways. Faseb J 14:2329-2338<br />

Balachandran P, Wei F, Lin RC, Khan IA, Pasco DS (2005) Structure activity relationships <strong>of</strong> aristolochic<br />

acid analogues: toxicity in cultured renal epithelial cells. <strong>Kidney</strong> Int 67:1797-1805<br />

Bannasch P (1986a) Preneoplastic lesions as end points in carcinogenicity testing. I. Hepatic<br />

preneoplasia. Carcinogenesis 7:689-695<br />

Bannasch P (1986b) Preneoplastic lesions as end points in carcinogenicity testing. II. Preneoplasia in<br />

various non-hepatic tissues. Carcinogenesis 7:849-852<br />

Bannasch P, Nogueira E, Zerban H (1989) [Cytology and cytogenesis <strong>of</strong> experimental epithelial kidney<br />

tumors]. Verh Dtsch Ges Pathol 73:301-313<br />

Barrett JC, Wiseman RW (1987) Cellular and molecular mechanisms <strong>of</strong> multistep carcinogenesis:<br />

relevance to carcinogen risk assessment. Environ Health Perspect 76:65-70<br />

Baugh LR, Hill AA, Brown EL, Hunter CP (2001) Quantitative analysis <strong>of</strong> mRNA amplification by in vitro<br />

transcription. Nucleic Acids Res 29:E29<br />

Baylin SB, Herman JG (2000) DNA hypermethylation in tumorigenesis: epigenetics joins genetics. Trends<br />

Genet 16:168-174<br />

Bellacosa A, Kumar CC, Di Crist<strong>of</strong>ano A, Testa JR (2005) Activation <strong>of</strong> AKT kinases in cancer:<br />

implications for therapeutic targeting. Adv <strong>Cancer</strong> Res 94:29-86<br />

Benesic A, Mildenberger S, Gekle M (2000) Nephritogenic ochratoxin A interferes with hormonal signalling<br />

in immortalized human kidney epithelial cells. Pflugers Arch 439:278-287<br />

Bergstrom A, Hsieh CC, Lindblad P, Lu CM, Cook NR, Wolk A (2001) Obesity and renal cell cancer--a<br />

quantitative review. Br J <strong>Cancer</strong> 85:984-990<br />

Bjornsson J, Short MP, Kwiatkowski DJ, Henske EP (1996) Tuberous sclerosis-associated renal cell<br />

carcinoma. Clinical, pathological, and genetic features. Am J Pathol 149:1201-1208<br />

Bonnet D, Dick JE (1997) Human acute myeloid leukemia is organized as a hierarchy that originates from<br />

a primitive hematopoietic cell. Nat Med 3:730-737<br />

Boorman GA, McDonald MR, Imoto S, Persing R (1992) Renal lesions induced by ochratoxin A exposure<br />

in the F344 rat. Toxicol Pathol 20:236-245<br />

188


Chapter 10: References<br />

Borenstein AR, Mortimer JA, Sch<strong>of</strong>ield E, Wu Y, Salmon DP, Gamst A, Olichney J, Thal LJ, Silbert L, Kaye<br />

J, Craig UL, Schellenberg GD, Galasko DR (2007) Cycad exposure and risk <strong>of</strong> dementia, MCI,<br />

and PDC in the Chamorro population <strong>of</strong> Guam. Neurology 68:1764-1771<br />

Branzei D, Foiani M (2008) Regulation <strong>of</strong> DNA repair throughout the cell cycle. Nat Rev Mol Cell Biol<br />

9:297-308<br />

Brugarolas JB, Vazquez F, Reddy A, Sellers WR, Kaelin WG, Jr. (2003) TSC2 regulates VEGF through<br />

mTOR-dependent and -independent pathways. <strong>Cancer</strong> Cell 4:147-158<br />

Casarett LJ, Doull J (1986) Toxic response <strong>of</strong> the kidney. Macmillan Publishing Company, New York<br />

Castaneda TR, Abplanalp W, Um S, Pfluger PT, Brown K, Grant E, Schrott E, Carnevalli L, Benoit SC,<br />

Morgan DA, Gilham D, Hui DY, Ramouhni K, Thomas G, Kozma SC, D.J. C, Tschöp MH (2008)<br />

The metabolic function <strong>of</strong> S6 kinases depends on dietary lipids. Cell Metab submitted<br />

Cavin C, Delatour T, Marin-Kuan M, Holzhauser D, Higgins L, Bezencon C, Guignard G, Junod S, Piguet<br />

D, Richoz-Payot J, Gremaux E, Hayes JD, Nestler S, Mantle P, Schilter B (2006) Reduction in<br />

Antioxidant Defences May Contribute to Ochratoxin A Toxicity and Carcinogenicity. Toxicol Sci<br />

Chan W, Cui L, Xu G, Cai Z (2006) Study <strong>of</strong> the phase I and phase II metabolism <strong>of</strong> nephrotoxin<br />

aristolochic acid by liquid chromatography/tandem mass spectrometry. Rapid Commun Mass<br />

Spectrom 20:1755-1760<br />

Chandra M, Riley MG, Johnson DE (1993) Spontaneous renal neoplasms in rats. J Appl Toxicol 13:109-<br />

116<br />

Chang HR, Lian JD, Lo CW, Chang YC, Yang MY, Wang CJ (2006) Induction <strong>of</strong> urothelial proliferation in<br />

rats by aristolochic acid through cell cycle progression via activation <strong>of</strong> cyclin D1/cdk4 and cyclin<br />

E/cdk2. Food Chem Toxicol 44:28-35<br />

Chen T, Guo L, Zhang L, Shi L, Fang H, Sun Y, Fuscoe JC, Mei N (2006) Gene Expression Pr<strong>of</strong>iles<br />

Distinguish the Carcinogenic Effects <strong>of</strong> Aristolochic Acid in Target (<strong>Kidney</strong>) and Non-target (Liver)<br />

Tissues in Rats. BMC Bioinformatics 7 Suppl 2:S20<br />

Cheng CL, Chen KJ, Shih PH, Lu LY, Hung CF, Lin WC, Yesong Gu J (2006) Chronic renal failure rats are<br />

highly sensitive to aristolochic acids, which are nephrotoxic and carcinogenic agents. <strong>Cancer</strong> Lett<br />

232:236-242<br />

Chipman JK, Mally A, Edwards GO (2003) Disruption <strong>of</strong> gap junctions in toxicity and carcinogenicity.<br />

Toxicol Sci 71:146-153<br />

Choi YJ, Kim CJ, Ji GE (1996) A partially purified beta-glucosidase from Bifidobacterium adolescentis<br />

converts cycasin to a mutagenic compound. Lett Appl Microbiol 22:145-148<br />

Chow WH, Gridley G, Fraumeni JF, Jr., Jarvholm B (2000) Obesity, hypertension, and the risk <strong>of</strong> kidney<br />

cancer in men. N Engl J Med 343:1305-1311<br />

Clark HA, Snedeker SM (2006) Ochratoxin a: its cancer risk and potential for exposure. J Toxicol Environ<br />

Health B Crit Rev 9:265-296<br />

Clarke MF, Dick JE, Dirks PB, Eaves CJ, Jamieson CH, Jones DL, Visvader J, Weissman IL, Wahl GM<br />

(2006) <strong>Cancer</strong> stem cells--perspectives on current status and future directions: AACR Workshop<br />

on cancer stem cells. <strong>Cancer</strong> Res 66:9339-9344<br />

Cohen SM, Arnold LL (2008) Cell Proliferation and Carcinogenesis. J Toxicol Pathol 21:1-7<br />

Cohen SM, Ellwein LB (1991) Genetic errors, cell proliferation, and carcinogenesis. <strong>Cancer</strong> Res 51:6493-<br />

6505<br />

Corgna E, Betti M, Gatta G, Roila F, De Mulder PH (2007) Renal cancer. Crit Rev Oncol Hematol 64:247-<br />

262<br />

Cosyns JP (2003) Aristolochic acid and 'Chinese herbs nephropathy': a review <strong>of</strong> the evidence to date.<br />

Drug Saf 26:33-48<br />

Cosyns JP, Goebbels RM, Liberton V, Schmeiser HH, Bieler CA, Bernard AM (1998) Chinese herbs<br />

nephropathy-associated slimming regimen induces tumours in the forestomach but no interstitial<br />

nephropathy in rats. Arch Toxicol 72:738-743<br />

Cosyns JP, Jadoul M, Squifflet JP, Wese FX, van Ypersele de Strihou C (1999) Urothelial lesions in<br />

Chinese-herb nephropathy. Am J <strong>Kidney</strong> Dis 33:1011-1017<br />

Cota D, Matter EK, Woods SC, Seeley RJ (2008) The role <strong>of</strong> hypothalamic mammalian target <strong>of</strong><br />

rapamycin complex 1 signaling in diet-induced obesity. J Neurosci 28:7202-7208<br />

Coughlan MT, Mibus AL, Forbes JM (2008) Oxidative stress and advanced glycation in diabetic<br />

nephropathy. Ann N Y Acad Sci 1126:190-193<br />

Crist<strong>of</strong>ori P, Zanetti E, Fregona D, Piaia A, Trevisan A (2007) Renal proximal tubule segment-specific<br />

nephrotoxicity: an overview on biomarkers and histopathology. Toxicol Pathol 35:270-275<br />

Cui M, Liu ZH, Qiu Q, Li H, Li LS (2005) Tumour induction in rats following exposure to short-term high<br />

dose aristolochic acid I. Mutagenesis 20:45-49<br />

Cunningham ML, Elwell MR, Matthews HB (1993) Site-specific cell proliferation in renal tubular cells by the<br />

renal tubular carcinogen tris(2,3-dibromopropyl)phosphate. Environ Health Perspect 101 Suppl<br />

5:253-257<br />

189


Chapter 10: References<br />

Davies TS, Lynch BS, Monro AM, Munro IC, Nestmann ER (2000) Rodent carcinogenicity tests need be<br />

no longer than 18 months: an analysis based on 210 chemicals in the IARC monographs. Food<br />

Chem Toxicol 38:219-235<br />

Debelle FD, Nortier JL, De Prez EG, Garbar CH, Vienne AR, Salmon IJ, Deschodt-Lanckman MM,<br />

Vanherweghem JL (2002) Aristolochic acids induce chronic renal failure with interstitial fibrosis in<br />

salt-depleted rats. J Am Soc Nephrol 13:431-436<br />

Delatour T, Mally A, Richoz J, Ozden S, Dekant W, Ihmels H, Otto D, Gasparutto D, Marin-Kuan M,<br />

Schilter B, Cavin C (2008) Absence <strong>of</strong> 2'-deoxyguanosine-carbon 8-bound ochratoxin A adduct in<br />

rat kidney DNA monitored by isotope dilution LC-MS/MS. Mol Nutr Food Res 52:472-482<br />

Delker D, Hatch G, Allen J, Crissman B, George M, Geter D, Kilburn S, Moore T, Nelson G, Roop B, Slade<br />

R, Swank A, Ward W, DeAngelo A (2006) <strong>Molecular</strong> biomarkers <strong>of</strong> oxidative stress associated<br />

with bromate carcinogenicity. Toxicology 221:158-165<br />

Dietrich DR, Swenberg JA (1990) Lindane induces nephropathy and renal accumulation <strong>of</strong> alpha 2uglobulin<br />

in male but not in female Fischer 344 rats or male NBR rats. Toxicol Lett 53:179-181<br />

Dietrich DR, Swenberg JA (1991) Preneoplastic lesions in rodent kidney induced spontaneously or by nongenotoxic<br />

agents: predictive nature and comparison to lesions induced by genotoxic carcinogens.<br />

Mutat Res 248:239-260<br />

Dill JA, Lee KM, Renne RA, Miller RA, Fuciarelli AF, Gideon KM, Chan PC, Burka LT, Roycr<strong>of</strong>t JH (2003)<br />

Alpha 2u-globulin nephropathy and carcinogenicity following exposure to decalin<br />

(decahydronaphthalene) in F344/N rats. Toxicol Sci 72:223-234<br />

Doi AM, Hill G, Seely J, Hailey JR, Kissling G, Bucher JR (2007) alpha 2u-globulin nephropathy and renal<br />

tumors in national toxicology program studies. Toxicol Pathol 35:533-540<br />

Domijan AM, Peraica M, Vrdoljak AL, Radic B, Zlender V, Fuchs R (2007) The involvement <strong>of</strong> oxidative<br />

stress in ochratoxin A and fumonisin B1 toxicity in rats. Mol Nutr Food Res 51:1147-1151<br />

Dong H, Suzuki N, Torres MC, Bonala RR, Johnson F, Grollman AP, Shibutani S (2006) Quantitative<br />

determination <strong>of</strong> aristolochic acid-derived DNA adducts in rats using 32Ppostlabeling/polyacrylamide<br />

gel electrophoresis analysis. Drug Metab Dispos 34:1122-1127<br />

Duarte CG, Preuss HG (1993) Assessment <strong>of</strong> renal function--glomerular and tubular. Clin Lab Med 13:33-<br />

52<br />

Dulaimi E, Ibanez de Caceres I, Uzzo RG, Al-Saleem T, Greenberg RE, Polascik TJ, Babb JS, Grizzle<br />

WE, Cairns P (2004) Promoter hypermethylation pr<strong>of</strong>ile <strong>of</strong> kidney cancer. Clin <strong>Cancer</strong> Res<br />

10:3972-3979<br />

Eddy AA, Fogo AB (2006) Plasminogen activator inhibitor-1 in chronic kidney disease: evidence and<br />

mechanisms <strong>of</strong> action. J Am Soc Nephrol 17:2999-3012<br />

Ellinger-Ziegelbauer H, Gmuender H, Bandenburg A, Ahr HJ (2008) Prediction <strong>of</strong> a carcinogenic potential<br />

<strong>of</strong> rat hepatocarcinogens using toxicogenomics analysis <strong>of</strong> short-term in vivo studies. Mutat Res<br />

637:23-39<br />

Ellinger-Ziegelbauer H, Stuart B, Wahle B, Bomann W, Ahr HJ (2004) Characteristic expression pr<strong>of</strong>iles<br />

induced by genotoxic carcinogens in rat liver. Toxicol Sci 77:19-34<br />

Ellinger-Ziegelbauer H, Stuart B, Wahle B, Bomann W, Ahr HJ (2005) Comparison <strong>of</strong> the expression<br />

pr<strong>of</strong>iles induced by genotoxic and nongenotoxic carcinogens in rat liver. Mutat Res 575:61-84<br />

Ellis M, Davis N, Coop A, Liu M, Schumaker L, Lee RY, Srikanchana R, Russell CG, Singh B, Miller WR,<br />

Stearns V, Pennanen M, Tsangaris T, Gallagher A, Liu A, Zwart A, Hayes DF, Lippman ME,<br />

Wang Y, Clarke R (2002) Development and validation <strong>of</strong> a method for using breast core needle<br />

biopsies for gene expression microarray analyses. Clin <strong>Cancer</strong> Res 8:1155-1166<br />

Emmert-Buck MR, Bonner RF, Smith PD, Chuaqui RF, Zhuang Z, Goldstein SR, Weiss RA, Liotta LA<br />

(1996) Laser capture microdissection. Science 274:998-1001<br />

Eustis SL (1989) The sequential development <strong>of</strong> cancer: a morphological perspective. Toxicol Lett 49:267-<br />

281<br />

Everitt JI, Goldsworthy TL, Wolf DC, Walker CL (1995) Hereditary renal cell carcinoma in the Eker rat: a<br />

unique animal model for the study <strong>of</strong> cancer susceptibility. Toxicol Lett 82-83:621-625<br />

Falvella FS, Colombo F, Spinola M, Campiglio M, Pastorino U, Dragani TA (2008) BHLHB3: a candidate<br />

tumor suppressor in lung cancer. Oncogene 27:3761-3764<br />

Faucet V, Pfohl-Leszkowicz A, Dai J, Castegnaro M, Manderville RA (2004) Evidence for covalent DNA<br />

adduction by ochratoxin A following chronic exposure to rat and subacute exposure to pig. Chem<br />

Res Toxicol 17:1289-1296<br />

Fearon ER, Vogelstein B (1990) A genetic model for colorectal tumorigenesis. Cell 61:759-767<br />

Fero ML, Randel E, Gurley KE, Roberts JM, Kemp CJ (1998) The murine gene p27Kip1 is haploinsufficient<br />

for tumour suppression. Nature 396:177-180<br />

Fiala ES, Bobotas G, Kulakis C, Weisburger JH (1976) Separation <strong>of</strong> 1,2-dimethylhydrazine metabolites by<br />

high-pressure liquid chromatography. J Chromatogr 117:181-185<br />

190


Chapter 10: References<br />

Fielden MR, Brennan R, Gollub J (2007) A gene expression biomarker provides early prediction and<br />

mechanistic assessment <strong>of</strong> hepatic tumor induction by nongenotoxic chemicals. Toxicol Sci<br />

99:90-100<br />

Flotow H, Thomas G (1992) Substrate recognition determinants <strong>of</strong> the mitogen-activated 70K S6 kinase<br />

from rat liver. J Biol Chem 267:3074-3078<br />

Foran JA (1997) Principles for the selection <strong>of</strong> doses in chronic rodent bioassays. ILSI Risk Science<br />

Working Group on Dose Selection. Environ Health Perspect 105:18-20<br />

Forbes JM, Coughlan MT, Cooper ME (2008) Oxidative stress as a major culprit in kidney disease in<br />

diabetes. Diabetes 57:1446-1454<br />

Fuchs R, Peraica M (2005) Ochratoxin A in human kidney diseases. Food Addit Contam 22 Suppl 1:53-57<br />

Fukunishi R, Watanabe K, Terashi S, Kawaji K (1971) Shift <strong>of</strong> target organs in cycasin carcinogenesis.<br />

Gann 62:353-358<br />

Furge KA, Dykema K, Petillo D, Westphal M, Zhang Z, Kort EJ, Teh BT (2007) Combining differential<br />

expression, chromosomal and pathway analyses for the molecular characterization <strong>of</strong> renal cell<br />

carcinoma. Can Urol Assoc J 1:S21-27<br />

Gagliano N, Torri C, Donetti E, Grizzi F, Costa F, Bertelli AA, Migliori M, Filippi C, Bedoni M, Panichi V,<br />

Giovannini L, Gioia M (2005) Ochratoxin A-induced renal cortex fibrosis and epithelial-tomesenchymal<br />

transition: molecular mechanisms <strong>of</strong> ochratoxin A-injury and potential effects <strong>of</strong> red<br />

wine. Mol Med 11:30-38<br />

Gekle M, Sauvant C, Schwerdt G (2005) Ochratoxin A at nanomolar concentrations: a signal modulator in<br />

renal cells. Mol Nutr Food Res 49:118-130<br />

Gekle M, Schwerdt G, Freudinger R, Mildenberger S, Wilflingseder D, Pollack V, Dander M, Schramek H<br />

(2000) Ochratoxin A induces JNK activation and apoptosis in MDCK-C7 cells at nanomolar<br />

concentrations. J Pharmacol Exp Ther 293:837-844<br />

Gekle M, Silbernagl S (1994) The role <strong>of</strong> the proximal tubule in ochratoxin A nephrotoxicity in vivo:<br />

toxodynamic and toxokinetic aspects. Ren Physiol Biochem 17:40-49<br />

Gold LI (1999) The role for transforming growth factor-beta (TGF-beta) in human cancer. Crit Rev Oncog<br />

10:303-360<br />

Goley EM, Anderson SJ, Menard C, Chuang E, Lu X, T<strong>of</strong>ilon PJ, Camphausen K (2004) Microarray<br />

analysis in clinical oncology: pre-clinical optimization using needle core biopsies from xenograft<br />

tumors. BMC <strong>Cancer</strong> 4:20<br />

Grab DJ, Zedeck MS (1977) Organ-specific effects <strong>of</strong> the carcinogen methylazoxymethanol related to<br />

metabolism by nicotinamide adenine dinucleotide-dependent dehydrogenases. <strong>Cancer</strong> Res<br />

37:4182-4189<br />

Grollman AP, Shibutani S, Moriya M, Miller F, Wu L, Moll U, Suzuki N, Fernandes A, Rosenquist T,<br />

Medverec Z, Jakovina K, Brdar B, Slade N, Turesky RJ, Goodenough AK, Rieger R, Vukelic M,<br />

Jelakovic B (2007) Aristolochic acid and the etiology <strong>of</strong> endemic (Balkan) nephropathy. Proc Natl<br />

Acad Sci U S A 104:12129-12134<br />

Gulati P, Gaspers LD, Dann SG, Joaquin M, Nobukuni T, Natt F, Kozma SC, Thomas AP, Thomas G<br />

(2008) Amino acids activate mTOR complex 1 via Ca2+/CaM signaling to hVps34. Cell Metab<br />

7:456-465<br />

Habib SL, Phan MN, Patel SK, Li D, Monks TJ, Lau SS (2003) Reduced constitutive 8-oxoguanine-DNA<br />

glycosylase expression and impaired induction following oxidative DNA damage in the tuberin<br />

deficient Eker rat. Carcinogenesis 24:573-582<br />

Hamadeh HK, Bushel PR, Jayadev S, Martin K, DiSorbo O, Sieber S, Bennett L, Tennant R, Stoll R,<br />

Barrett JC, Blanchard K, Paules RS, Afshari CA (2002) Gene expression analysis reveals<br />

chemical-specific pr<strong>of</strong>iles. Toxicol Sci 67:219-231<br />

Hamilton JM (1975) Renal carcinogenesis. Adv <strong>Cancer</strong> Res 22:1-56<br />

Hanahan D, Weinberg RA (2000) The hallmarks <strong>of</strong> cancer. Cell 100:57-70<br />

Hanna SC, Heathcote SA, Kim WY (2008) mTOR pathway in renal cell carcinoma. Expert Rev Anticancer<br />

Ther 8:283-292<br />

Hard GC (1998) Mechanisms <strong>of</strong> chemically induced renal carcinogenesis in the laboratory rodent. Toxicol<br />

Pathol 26:104-112<br />

Hard GC, Khan KN (2004) A contemporary overview <strong>of</strong> chronic progressive nephropathy in the laboratory<br />

rat, and its significance for human risk assessment. Toxicol Pathol 32:171-180<br />

Harrington LS, Findlay GM, Gray A, Tolkacheva T, Wigfield S, Rebholz H, Barnett J, Leslie NR, Cheng S,<br />

Shepherd PR, Gout I, Downes CP, Lamb RF (2004) The TSC1-2 tumor suppressor controls<br />

insulin-PI3K signaling via regulation <strong>of</strong> IRS proteins. J Cell Biol 166:213-223<br />

Haseman J, Melnick R, Tomatis L, Huff J (2001) Carcinogenesis bioassays: study duration and biological<br />

relevance. Food Chem Toxicol 39:739-744<br />

Hengstler JG, Bogdanffy MS, Bolt HM, Oesch F (2003) Challenging dogma: thresholds for genotoxic<br />

carcinogens? The case <strong>of</strong> vinyl acetate. Annu Rev Pharmacol Toxicol 43:485-520<br />

191


Chapter 10: References<br />

Henske EP (2005) Tuberous sclerosis and the kidney: from mesenchyme to epithelium, and beyond.<br />

Pediatr Nephrol 20:854-857<br />

Hino O (2004) Multistep renal carcinogenesis in the Eker (Tsc 2 gene mutant) rat model. Curr Mol Med<br />

4:807-811<br />

Hino O, Kobayashi E, Hirayama Y, Kobayashi T, Kubo Y, Tsuchiya H, Kikuchi Y, Mitani H (1995)<br />

<strong>Molecular</strong> genetic basis <strong>of</strong> renal carcinogenesis in the Eker rat model <strong>of</strong> tuberous sclerosis<br />

(Tsc2). Mol Carcinog 14:23-27<br />

Hino O, Mitani H, Knudson AG (1993) Genetic predisposition to transplacentally induced renal cell<br />

carcinomas in the Eker rat. <strong>Cancer</strong> Res 53:5856-5858<br />

Hirono I (1972) Carcinogenicity and neurotoxicity <strong>of</strong> cycasin with special reference to species differences.<br />

Fed Proc 31:1493-1499<br />

Hirono I (1981) Natural carcinogenic products <strong>of</strong> plant origin. Crit Rev Toxicol 8:235-277<br />

Hirono I, Laqueur GL, Spatz M (1968) Tumor induction in Fischer and Osborne-Mendel rats by a single<br />

administration <strong>of</strong> cycasin. J Natl <strong>Cancer</strong> Inst 40:1003-1010<br />

Hjartaker A, Langseth H, Weiderpass E (2008) Obesity and diabetes epidemics: cancer repercussions.<br />

Adv Exp Med Biol 630:72-93<br />

Horiguchi A, Sumitomo M, Asakuma J, Asano T, Zheng R, Asano T, Nanus DM, Hayakawa M (2006)<br />

Increased serum leptin levels and over expression <strong>of</strong> leptin receptors are associated with the<br />

invasion and progression <strong>of</strong> renal cell carcinoma. J Urol 176:1631-1635<br />

Horvath A, Upham BL, Ganev V, Trosko JE (2002) Determination <strong>of</strong> the epigenetic effects <strong>of</strong> ochratoxin in<br />

a human kidney and a rat liver epithelial cell line. Toxicon 40:273-282<br />

Hotamisligil GS (2006) Inflammation and metabolic disorders. Nature 444:860-867<br />

Howard PC, Eppley RM, Stack ME, Warbritton A, Voss KA, Lorentzen RJ, Kovach RM, Bucci TJ (2001)<br />

Fumonisin b1 carcinogenicity in a two-year feeding study using F344 rats and B6C3F1 mice.<br />

Environ Health Perspect 109 Suppl 2:277-282<br />

Hussain SP, Harris CC (1998) <strong>Molecular</strong> epidemiology <strong>of</strong> human cancer: contribution <strong>of</strong> mutation spectra<br />

studies <strong>of</strong> tumor suppressor genes. <strong>Cancer</strong> Res 58:4023-4037<br />

IARC (2002a) International Agency for Reasearch on <strong>Cancer</strong>. IARC Handbooks <strong>of</strong> <strong>Cancer</strong> Prevention.<br />

Weight Control and Physical Activity. Lyon.<br />

IARC (2002b) International Agency for Research on <strong>Cancer</strong>. Some traditional herbal medicines, some<br />

mycotoxins, naphthalene and styrene. IARC Monogr. Eval. Carcinog. Risks Hum. 82<br />

IARC (2002c) International Agency For Research on <strong>Cancer</strong>: <strong>Cancer</strong> Incidence in Five Continents, Vol.<br />

VIII. Scientific Publication No. 155<br />

ICH (1997) International Conference on Harmonization <strong>of</strong> Technical Requirenments for the Registration on<br />

Pharmaceutical for Human Use. Harmonized Guideline. testing for Carcinogenicity <strong>of</strong><br />

Pharmaceuticals. CPMP/ICH/299/95<br />

Ince PG, Codd GA (2005) Return <strong>of</strong> the cycad hypothesis - does the amyotrophic lateral<br />

sclerosis/parkinsonism dementia complex (ALS/PDC) <strong>of</strong> Guam have new implications for global<br />

health? Neuropathol Appl Neurobiol 31:345-353<br />

Jacinto E, Facchinetti V, Liu D, Soto N, Wei S, Jung SY, Huang Q, Qin J, Su B (2006) SIN1/MIP1<br />

maintains rictor-mTOR complex integrity and regulates Akt phosphorylation and substrate<br />

specificity. Cell 127:125-137<br />

Jacinto E, Loewith R, Schmidt A, Lin S, Ruegg MA, Hall A, Hall MN (2004) Mammalian TOR complex 2<br />

controls the actin cytoskeleton and is rapamycin insensitive. Nat Cell Biol 6:1122-1128<br />

Jaffe AB, Hall A (2002) Rho GTPases in transformation and metastasis. Adv <strong>Cancer</strong> Res 84:57-80<br />

Jefford CE, Irminger-Finger I (2006) Mechanisms <strong>of</strong> chromosome instability in cancers. Crit Rev Oncol<br />

Hematol 59:1-14<br />

Jiang T, Wang Z, Proctor G, Moskowitz S, Liebman SE, Rogers T, Lucia MS, Li J, Levi M (2005) Dietinduced<br />

obesity in C57BL/6J mice causes increased renal lipid accumulation and<br />

glomerulosclerosis via a sterol regulatory element-binding protein-1c-dependent pathway. J Biol<br />

Chem 280:32317-32325<br />

Jones J, Pantuck AJ (2008) Genomics and proteomics in renal cell carcinoma: diagnosis, prognosis, and<br />

treatment selection. Curr Urol Rep 9:9-14<br />

Jozwiak J (2006) Hamartin and tuberin: working together for tumour suppression. Int J <strong>Cancer</strong> 118:1-5<br />

Kaimori JY, Takenaka M, Nakajima H, Hamano T, Horio M, Sugaya T, Ito T, Hori M, Okubo K, Imai E<br />

(2003) Induction <strong>of</strong> glia maturation factor-beta in proximal tubular cells leads to vulnerability to<br />

oxidative injury through the p38 pathway and changes in antioxidant enzyme activities. J Biol<br />

Chem 278:33519-33527<br />

Kamp HG, Eisenbrand G, Janzowski C, Kiossev J, Latendresse JR, Schlatter J, Turesky RJ (2005)<br />

Ochratoxin A induces oxidative DNA damage in liver and kidney after oral dosing to rats. Mol Nutr<br />

Food Res 49:1160-1167<br />

192


Chapter 10: References<br />

Keenan KP, Laroque P, Ballam GC, Soper KA, Dixit R, Mattson BA, Adams SP, Coleman JB (1996) The<br />

effects <strong>of</strong> diet, ad libitum overfeeding, and moderate dietary restriction on the rodent bioassay:<br />

the uncontrolled variable in safety assessment. Toxicol Pathol 24:757-768<br />

Keenan KP, Soper KA, Smith PF, Ballam GC, Clark RL (1995) Diet, overfeeding, and moderate dietary<br />

restriction in control Sprague-Dawley rats: I. Effects on spontaneous neoplasms. Toxicol Pathol<br />

23:269-286<br />

Kenerson H, Dundon TA, Yeung RS (2005) Effects <strong>of</strong> rapamycin in the Eker rat model <strong>of</strong> tuberous<br />

sclerosis complex. Pediatr Res 57:67-75<br />

Kenerson HL, Aicher LD, True LD, Yeung RS (2002) Activated mammalian target <strong>of</strong> rapamycin pathway in<br />

the pathogenesis <strong>of</strong> tuberous sclerosis complex renal tumors. <strong>Cancer</strong> Res 62:5645-5650<br />

Kimura H, Odani S, Suzuki J, Arakawa M, Ono T (1989) <strong>Kidney</strong> fatty acid-binding protein: identification as<br />

alpha 2U-globulin. FEBS Lett 246:101-104<br />

Klaunig JE, Kamendulis LM (2004) The role <strong>of</strong> oxidative stress in carcinogenesis. Annu Rev Pharmacol<br />

Toxicol 44:239-267<br />

Knudson AG, Jr. (1975) Genetics <strong>of</strong> human cancer. Genetics 79 Suppl:305-316<br />

Kobayashi T, Hirayama Y, Kobayashi E, Kubo Y, Hino O (1995) A germline insertion in the tuberous<br />

sclerosis (Tsc2) gene gives rise to the Eker rat model <strong>of</strong> dominantly inherited cancer. Nat Genet<br />

9:70-74<br />

Kobayashi T, Urakami S, Hirayama Y, Yamamoto T, Nishizawa M, Takahara T, Kubo Y, Hino O (1997)<br />

Intragenic Tsc2 somatic mutations as Knudson's second hit in spontaneous and chemically<br />

induced renal carcinomas in the Eker rat model. Jpn J <strong>Cancer</strong> Res 88:254-261<br />

Kohara A, Suzuki T, Honma M, Ohwada T, Hayashi M (2002) Mutagenicity <strong>of</strong> aristolochic acid in the<br />

lambda/lacZ transgenic mouse (MutaMouse). Mutat Res 515:63-72<br />

Kovacs G, Akhtar M, Beckwith BJ, Bugert P, Cooper CS, Delahunt B, Eble JN, Fleming S, Ljungberg B,<br />

Medeiros LJ, Moch H, Reuter VE, Ritz E, Roos G, Schmidt D, Srigley JR, Storkel S, van den Berg<br />

E, Zbar B (1997) The Heidelberg classification <strong>of</strong> renal cell tumours. J Pathol 183:131-133<br />

Krumbiegel G, Hallensleben J, Mennicke WH, Rittmann N, Roth HJ (1987) Studies on the metabolism <strong>of</strong><br />

aristolochic acids I and II. Xenobiotica 17:981-991<br />

Kubo Y, Mitani H, Hino O (1994) Allelic loss at the predisposing gene locus in spontaneous and chemically<br />

induced renal cell carcinomas in the Eker rat. <strong>Cancer</strong> Res 54:2633-2635<br />

Kuiper-Goodman T (1991) Risk assessment <strong>of</strong> ochratoxin A residues in food. IARC Sci Publ:307-320<br />

Kwiatkowski DJ (2003) Rhebbing up mTOR: new insights on TSC1 and TSC2, and the pathogenesis <strong>of</strong><br />

tuberous sclerosis. <strong>Cancer</strong> Biol Ther 2:471-476<br />

Laping NJ, Everitt JI, Frazier KS, Burgert M, Portis MJ, Cadacio C, Gold LI, Walker CL (2007) Tumorspecific<br />

efficacy <strong>of</strong> transforming growth factor-beta RI inhibition in Eker rats. Clin <strong>Cancer</strong> Res<br />

13:3087-3099<br />

Laqueur GL (1964) Carcinogenic Effects Of Cycad Meal And Cycasin, Methylazoxymethanol Glycoside, In<br />

Rats And Effects Of Cycasin In Germfree Rats. Fed Proc 23:1386-1388<br />

Laqueur GL (1968) Toxicology <strong>of</strong> cycasin. Food Cosmet Toxicol 6:577-578<br />

Laqueur GL, Matsumoto H (1966) Neoplasms in female Fischer rats following intraperitoneal injection <strong>of</strong><br />

methylazoxy-methanol. J Natl <strong>Cancer</strong> Inst 37:217-232<br />

Lawrence MC, Jivan A, Shao C, Duan L, Goad D, Zaganjor E, Osborne J, McGlynn K, Stippec S, Earnest<br />

S, Chen W, Cobb MH (2008) The roles <strong>of</strong> MAPKs in disease. Cell Res 18:436-442<br />

Le Hir M, Dubach UC (1982) Peroxisomal and mitochondrial beta-oxidation in the rat kidney: distribution <strong>of</strong><br />

fatty acyl-coenzyme A oxidase and 3-hydroxyacyl-coenzyme A dehydrogenase activities along<br />

the nephron. J Histochem Cytochem 30:441-444<br />

Leighton JK, Brown P, Ellis A, Harlow P, Harrouk W, Pine PS, Robison T, Rosario L, Thompson K (2006)<br />

Workgroup report: Review <strong>of</strong> genomics data based on experience with mock submissions--view<br />

<strong>of</strong> the CDER Pharmacology Toxicology Nonclinical Pharmacogenomics Subcommittee. Environ<br />

Health Perspect 114:573-578<br />

Leithe E, Sirnes S, Omori Y, Rivedal E (2006) Downregulation <strong>of</strong> gap junctions in cancer cells. Crit Rev<br />

Oncog 12:225-256<br />

Leivonen SK, Kahari VM (2007) Transforming growth factor-beta signaling in cancer invasion and<br />

metastasis. Int J <strong>Cancer</strong> 121:2119-2124<br />

Levin BE, Triscari J, Hogan S, Sullivan AC (1987) Resistance to diet-induced obesity: food intake,<br />

pancreatic sympathetic tone, and insulin. Am J Physiol 252:R471-478<br />

Li CM, Guo M, Borczuk A, Powell CA, Wei M, Thaker HM, Friedman R, Klein U, Tycko B (2002) Gene<br />

expression in Wilms' tumor mimics the earliest committed stage in the metanephric<br />

mesenchymal-epithelial transition. Am J Pathol 160:2181-2190<br />

Li Y, Liu Z, Guo X, Shu J, Chen Z, Li L (2006) Aristolochic acid I-induced DNA damage and cell cycle<br />

arrest in renal tubular epithelial cells in vitro. Arch Toxicol 80:524-532<br />

Liang J, Slingerland JM (2003) Multiple roles <strong>of</strong> the PI3K/PKB (Akt) pathway in cell cycle progression. Cell<br />

Cycle 2:339-345<br />

193


Chapter 10: References<br />

Lijinsky W, Kovatch RM, Riggs CW, Walters PT (1988) Dose-response study with N-nitrosomorpholine in<br />

drinking water <strong>of</strong> F-344 rats. <strong>Cancer</strong> Res 48:2089-2095<br />

Lina BA, Kuijpers MH (2004) Toxicity and carcinogenicity <strong>of</strong> acidogenic or alkalogenic diets in rats; effects<br />

<strong>of</strong> feeding NH(4)Cl, KHCO(3) or KCl. Food Chem Toxicol 42:135-153<br />

Liu MC, Maruyama S, Mizuno M, Morita Y, Hanaki S, Yuzawa Y, Matsuo S (2003) The nephrotoxicity <strong>of</strong><br />

Aristolochia manshuriensis in rats is attributable to its aristolochic acids. Clin Exp Nephrol 7:186-<br />

194<br />

Lock EA, Hard GC (2004) Chemically induced renal tubule tumors in the laboratory rat and mouse: review<br />

<strong>of</strong> the NCI/NTP database and categorization <strong>of</strong> renal carcinogens based on mechanistic<br />

information. Crit Rev Toxicol 34:211-299<br />

Lord GM, Hollstein M, Arlt VM, Roufosse C, Pusey CD, Cook T, Schmeiser HH (2004) DNA adducts and<br />

p53 mutations in a patient with aristolochic acid-associated nephropathy. Am J <strong>Kidney</strong> Dis<br />

43:e11-17<br />

Luhe A, Hildebrand H, Bach U, Dingermann T, Ahr HJ (2003) A new approach to studying ochratoxin A<br />

(OTA)-induced nephrotoxicity: expression pr<strong>of</strong>iling in vivo and in vitro employing cDNA<br />

microarrays. Toxicol Sci 73:315-328<br />

Luhe A, Suter L, Ruepp S, Singer T, Weiser T, Albertini S (2005) Toxicogenomics in the pharmaceutical<br />

industry: hollow promises or real benefit? Mutat Res 575:102-115<br />

Ma L, Chen Z, Erdjument-Bromage H, Tempst P, Pandolfi PP (2005a) Phosphorylation and functional<br />

inactivation <strong>of</strong> TSC2 by Erk implications for tuberous sclerosis and cancer pathogenesis. Cell<br />

121:179-193<br />

Ma L, Teruya-Feldstein J, Behrendt N, Chen Z, Noda T, Hino O, Cordon-Cardo C, Pandolfi PP (2005b)<br />

Genetic analysis <strong>of</strong> Pten and Tsc2 functional interactions in the mouse reveals asymmetrical<br />

haploinsufficiency in tumor suppression. Genes Dev 19:1779-1786<br />

Mak BC, Yeung RS (2004) The tuberous sclerosis complex genes in tumor development. <strong>Cancer</strong> Invest<br />

22:588-603<br />

Mally A, Pepe G, Ravoori S, Fiore M, Gupta RC, Dekant W, Mosesso P (2005) Ochratoxin a causes DNA<br />

damage and cytogenetic effects but no DNA adducts in rats. Chem Res Toxicol 18:1253-1261<br />

Malumbres M, Barbacid M (2001) To cycle or not to cycle: a critical decision in cancer. Nat Rev <strong>Cancer</strong><br />

1:222-231<br />

Manning BD, Cantley LC (2007) AKT/PKB signaling: navigating downstream. Cell 129:1261-1274<br />

Manning BD, Logsdon MN, Lipovsky AI, Abbott D, Kwiatkowski DJ, Cantley LC (2005) Feedback inhibition<br />

<strong>of</strong> Akt signaling limits the growth <strong>of</strong> tumors lacking Tsc2. Genes Dev 19:1773-1778<br />

Mantle P, Kulinskaya E, Nestler S (2005) Renal tumourigenesis in male rats in response to chronic dietary<br />

ochratoxin A. Food Addit Contam 22 Suppl 1:58-64<br />

Marin-Kuan M, Nestler S, Verguet C, Bezencon C, Piguet D, Delatour T, Mantle P, Cavin C, Schilter B<br />

(2007) MAPK-ERK activation in kidney <strong>of</strong> male rats chronically fed ochratoxin A at a dose<br />

causing a significant incidence <strong>of</strong> renal carcinoma. Toxicol Appl Pharmacol 224:174-181<br />

Marin-Kuan M, Nestler S, Verguet C, Bezencon C, Piguet D, Mansourian R, Holzwarth J, Grigorov M,<br />

Delatour T, Mantle P, Cavin C, Schilter B (2006) A toxicogenomics approach to identify new<br />

plausible epigenetic mechanisms <strong>of</strong> ochratoxin a carcinogenicity in rat. Toxicol Sci 89:120-134<br />

Martinez-Iglesias O, Ruiz-Llorente L, Sanchez-Martinez R, Garcia L, Zambrano A, Aranda A (2008)<br />

Histone deacetylase inhibitors: mechanism <strong>of</strong> action and therapeutic use in cancer. Clin Transl<br />

Oncol 10:395-398<br />

Matsumoto H, Higa HH (1966) Studies on methylazoxymethanol, the aglycone <strong>of</strong> cycasin: methylation <strong>of</strong><br />

nucleic acids in vitro. Biochem J 98:20C-22C<br />

Matsushima T, Matsumoto H, Shirai A, Sawamura M, Sugimura T (1979) Mutagenicity <strong>of</strong> the naturally<br />

occurring carcinogen cycasin and synthetic methylazoxymethanol conjugates in Salmonella<br />

typhimurium. <strong>Cancer</strong> Res 39:3780-3782<br />

Mayer D, Weber E, Kadenbach B, Bannasch P (1989) Immunocytochemical demonstration <strong>of</strong> cytochrome<br />

c oxidase as a marker for renal oncocytes and oncocytomas. Toxicol Pathol 17:46-49<br />

McDorman KS, Wolf DC (2002) Use <strong>of</strong> the spontaneous Tsc2 knockout (Eker) rat model <strong>of</strong> hereditary<br />

renal cell carcinoma for the study <strong>of</strong> renal carcinogens. Toxicol Pathol 30:675-680<br />

McLaughlin JK, Lipworth L (2000) Epidemiologic aspects <strong>of</strong> renal cell cancer. Semin Oncol 27:115-123<br />

McTiernan A (2005) Obesity and cancer: the risks, science, and potential management strategies.<br />

Oncology (Williston Park) 19:871-881; discussion 881-872, 885-876<br />

Mei N, Arlt VM, Phillips DH, Heflich RH, Chen T (2006) DNA adduct formation and mutation induction by<br />

aristolochic acid in rat kidney and liver. Mutat Res 602:83-91<br />

Mellemgaard A, Lindblad P, Schleh<strong>of</strong>er B, Bergstrom R, Mandel JS, McCredie M, McLaughlin JK, Niwa S,<br />

Odaka N, Pommer W, et al. (1995) International renal-cell cancer study. III. Role <strong>of</strong> weight,<br />

height, physical activity, and use <strong>of</strong> amphetamines. Int J <strong>Cancer</strong> 60:350-354<br />

Mengs U (1987) Acute toxicity <strong>of</strong> aristolochic acid in rodents. Arch Toxicol 59:328-331<br />

194


Chapter 10: References<br />

Mengs U, Lang W, Poch J-A (1982) The Carcinogenic Action <strong>of</strong> Aristolochic Acid in Rats. Archives <strong>of</strong><br />

Toxicology 51:107-119<br />

Mengs U, Stotzem CD (1993) Renal toxicity <strong>of</strong> aristolochic acid in rats as an example <strong>of</strong> nephrotoxicity<br />

testing in routine toxicology. Arch Toxicol 67:307-311<br />

Momparler RL (2003) <strong>Cancer</strong> epigenetics. Oncogene 22:6479-6483<br />

Monro A (1993) How useful are chronic (life-span) toxicology studies in rodents in identifying<br />

pharmaceuticals that pose a carcinogenic risk to humans? Adverse Drug React Toxicol Rev 12:5-<br />

34<br />

Moore B, Beltran L, Carbajal S, Strom S, Traag J, Hursting SD, DiGiovanni J (2008) Dietary Energy<br />

Balance Modulates Signaling through the Akt/Mammalian Target <strong>of</strong> Rapamycin Pathways in Multiple<br />

Epithelial Tissues. <strong>Cancer</strong> Prev Res June:65-76<br />

Morton LD, Youssef AF, Lloyd E, Kiorpes AL, Goldsworthy TL, Fort FL (2002) Evaluation <strong>of</strong> carcinogenic<br />

responses in the Eker rat following short-term exposure to selected nephrotoxins and<br />

carcinogens. Toxicol Pathol 30:559-564<br />

Motzer RJ, Bander NH, Nanus DM (1996) Renal-cell carcinoma. N Engl J Med 335:865-875<br />

Nagasawa HT, Shirota FN, Matsumoto H (1972) Decomposition <strong>of</strong> methylazoxymethanol, the aglycone <strong>of</strong><br />

cycasin, in D 2 O. Nature 236:234-235<br />

Nogueira E (1987) Rat renal carcinogenesis after chronic simultaneous exposure to lead acetate and Nnitrosodiethylamine.<br />

Virchows Arch B Cell Pathol Incl Mol Pathol 53:365-374<br />

Nogueira E, Klimek F, Weber E, Bannasch P (1989) Collecting duct origin <strong>of</strong> rat renal clear cell tumors.<br />

Virchows Arch B Cell Pathol Incl Mol Pathol 57:275-283<br />

Nortier JL, Martinez MC, Schmeiser HH, Arlt VM, Bieler CA, Petein M, Depierreux MF, De Pauw L,<br />

Abramowicz D, Vereerstraeten P, Vanherweghem JL (2000) Urothelial carcinoma associated with<br />

the use <strong>of</strong> a Chinese herb (Aristolochia fangchi). N Engl J Med 342:1686-1692<br />

Nortier JL, Vanherweghem JL (2002) Renal interstitial fibrosis and urothelial carcinoma associated with the<br />

use <strong>of</strong> a Chinese herb (Aristolochia fangchi). Toxicology 181-182:577-580<br />

Notman J, Tan QH, Zedeck MS (1982) Inhibition <strong>of</strong> methylazoxymethanol-induced intestinal tumors in the<br />

rat by pyrazole with paradoxical effects on skin and kidney. <strong>Cancer</strong> Res 42:1774-1780<br />

NTP (1989) Toxicology and Carcinogenesis Studies <strong>of</strong> Ochratoxin A (CAS No. 303-47-9) in F344/N Rats<br />

(Gavage Studies). Natl Toxicol Program Tech Rep Ser 358:1-142<br />

NTP US (1992) U.S. NTP. Specification for the Conduct <strong>of</strong> Studies to Evaluate the Toxic and Carcinogenic<br />

Potential <strong>of</strong> Chemical, Biological and Physical Agents in Laboratory Animals for the National Toxicology<br />

Program (NTP).<br />

Nunez NP, Oh WJ, Rozenberg J, Perella C, Anver M, Barrett JC, Perkins SN, Berrigan D, Moitra J,<br />

Varticovski L, Hursting SD, Vinson C (2006) Accelerated tumor formation in a fatless mouse with<br />

type 2 diabetes and inflammation. <strong>Cancer</strong> Res 66:5469-5476<br />

Nygaard V, Holden M, Loland A, Langaas M, Myklebost O, Hovig E (2005) Limitations <strong>of</strong> mRNA<br />

amplification from small-size cell samples. BMC Genomics 6:147<br />

O'Brien CA, Pollett A, Gallinger S, Dick JE (2007) A human colon cancer cell capable <strong>of</strong> initiating tumour<br />

growth in immunodeficient mice. Nature 445:106-110<br />

O'Brien E, Dietrich DR (2005) Ochratoxin A: the continuing enigma. Crit Rev Toxicol 35:33-60<br />

O'Gara RW, Brown JM, Whiting MG (1964) Induction Of Hepatic And Renal Tumors By Topical Application<br />

Of Aqueous Extract Of Cycad Nut To Artificial Skin Ulcers In Mice. Fed Proc 23:1382-1383<br />

OECD (1981) OECD guidelines for testing <strong>of</strong> chemicals. Paris:Organisation for Economic Co-operation<br />

and Development.<br />

Oesch F, Herrero ME, Hengstler JG, Lohmann M, Arand M (2000) Metabolic detoxification: implications for<br />

thresholds. Toxicol Pathol 28:382-387<br />

Ogden CL, Yanovski SZ, Carroll MD, Flegal KM (2007) The epidemiology <strong>of</strong> obesity. Gastroenterology<br />

132:2087-2102<br />

Ono H, Pocai A, Wang Y, Sakoda H, Asano T, Backer JM, Schwartz GJ, Rossetti L (2008) Activation <strong>of</strong><br />

hypothalamic S6 kinase mediates diet-induced hepatic insulin resistance in rats. J Clin Invest<br />

118:2959-2968<br />

Pantuck AJ, Thomas G, Belldegrun AS, Figlin RA (2006) Mammalian target <strong>of</strong> rapamycin inhibitors in renal<br />

cell carcinoma: current status and future applications. Semin Oncol 33:607-613<br />

Parsa AT, Waldron JS, Panner A, Crane CA, Parney IF, Barry JJ, Cachola KE, Murray JC, Tihan T,<br />

Jensen MC, Mischel PS, Stokoe D, Pieper RO (2007) Loss <strong>of</strong> tumor suppressor PTEN function<br />

increases B7-H1 expression and immunoresistance in glioma. Nat Med 13:84-88<br />

Pavlovich CP, Schmidt LS (2004) Searching for the hereditary causes <strong>of</strong> renal-cell carcinoma. Nat Rev<br />

<strong>Cancer</strong> 4:381-393<br />

Perona R (2006) Cell signalling: growth factors and tyrosine kinase receptors. Clin Transl Oncol 8:77-82<br />

Peto R, Gray R, Brantom P, Grasso P (1991) Dose and time relationships for tumor induction in the liver<br />

and esophagus <strong>of</strong> 4080 inbred rats by chronic ingestion <strong>of</strong> N-nitrosodiethylamine or Nnitrosodimethylamine.<br />

<strong>Cancer</strong> Res 51:6452-6469<br />

195


Chapter 10: References<br />

Pfau W, Schmeiser HH, Wiessler M (1990) Aristolochic acid binds covalently to the exocyclic amino group<br />

<strong>of</strong> purine nucleotides in DNA. Carcinogenesis 11:313-319<br />

Pfau W, Schmeiser HH, Wiessler M (1991) N6-adenyl arylation <strong>of</strong> DNA by aristolochic acid II and a<br />

synthetic model for the putative proximate carcinogen. Chem Res Toxicol 4:581-586<br />

Rached E, Pfeiffer E, Dekant W, Mally A (2006) Ochratoxin A: apoptosis and aberrant exit from mitosis<br />

due to perturbation <strong>of</strong> microtubule dynamics? Toxicol Sci 92:78-86<br />

Radulovic S, Bjelogrlic SK (2007) Sunitinib, sorafenib and mTOR inhibitors in renal cancer. J Buon 12<br />

Suppl 1:S151-162<br />

Raghavan N, Amaratunga D, Nie AY, McMillian M (2005) Class prediction in toxicogenomics. J Biopharm<br />

Stat 15:327-341<br />

Rahimtula AD, Chong X (1991) Alterations in calcium homeostasis as a possible cause <strong>of</strong> ochratoxin A<br />

nephrotoxicity. IARC Sci Publ:207-214<br />

Rasonyi T, Schlatter J, Dietrich DR (1999) The role <strong>of</strong> alpha2u-globulin in ochratoxin A induced renal<br />

toxicity and tumors in F344 rats. Toxicol Lett 104:83-92<br />

Ravitz MJ, Chen L, Lynch M, Schmidt EV (2007) c-myc Repression <strong>of</strong> TSC2 contributes to control <strong>of</strong><br />

translation initiation and Myc-induced transformation. <strong>Cancer</strong> Res 67:11209-11217<br />

Ravussin E (2005) Physiology. A NEAT way to control weight? Science 307:530-531<br />

Ravussin E, Danforth E, Jr. (1999) Beyond sloth--physical activity and weight gain. Science 283:184-185<br />

Renehan AG, Frystyk J, Flyvbjerg A (2006) Obesity and cancer risk: the role <strong>of</strong> the insulin-IGF axis.<br />

Trends Endocrinol Metab 17:328-336<br />

Renehan AG, Roberts DL, Dive C (2008) Obesity and cancer: pathophysiological and biological<br />

mechanisms. Arch Physiol Biochem 114:71-83<br />

Rennebeck G, Kleymenova EV, Anderson R, Yeung RS, Artzt K, Walker CL (1998) Loss <strong>of</strong> function <strong>of</strong> the<br />

tuberous sclerosis 2 tumor suppressor gene results in embryonic lethality characterized by<br />

disrupted neuroepithelial growth and development. Proc Natl Acad Sci U S A 95:15629-15634<br />

Rockett JC, Hellmann GM (2004) Confirming microarray data-is it really necessary? Genomics 83:541-549<br />

Rosner M, H<strong>of</strong>er K, Kubista M, Hengstschlager M (2003) Cell size regulation by the human TSC tumor<br />

suppressor proteins depends on PI3K and FKBP38. Oncogene 22:4786-4798<br />

Sarbassov DD, Ali SM, Kim DH, Guertin DA, Latek RR, Erdjument-Bromage H, Tempst P, Sabatini DM<br />

(2004) Rictor, a novel binding partner <strong>of</strong> mTOR, defines a rapamycin-insensitive and raptorindependent<br />

pathway that regulates the cytoskeleton. Curr Biol 14:1296-1302<br />

Sarbassov DD, Guertin DA, Ali SM, Sabatini DM (2005) Phosphorylation and regulation <strong>of</strong> Akt/PKB by the<br />

rictor-mTOR complex. Science 307:1098-1101<br />

Sauvant C, Holzinger H, Mildenberger S, Gekle M (2005) Exposure to nephrotoxic ochratoxin A enhances<br />

collagen secretion in human renal proximal tubular cells. Mol Nutr Food Res 49:31-37<br />

Schmeiser HH, Janssen JW, Lyons J, Scherf HR, Pfau W, Buchmann A, Bartram CR, Wiessler M (1990)<br />

Aristolochic acid activates ras genes in rat tumors at deoxyadenosine residues. <strong>Cancer</strong> Res<br />

50:5464-5469<br />

Schmeiser HH, Pool BL, Wiessler M (1984) Mutagenicity <strong>of</strong> the two main components <strong>of</strong> commercially<br />

available carcinogenic aristolochic acid in Salmonella typhimurium. <strong>Cancer</strong> Lett 23:97-101<br />

Schmeiser HH, Scherf HR, Wiessler M (1991) Activating mutations at codon 61 <strong>of</strong> the c-Ha-ras gene in<br />

thin-tissue sections <strong>of</strong> tumors induced by aristolochic acid in rats and mice. <strong>Cancer</strong> Lett 59:139-<br />

143<br />

Schmeiser HH, Schoepe KB, Wiessler M (1988) DNA adduct formation <strong>of</strong> aristolochic acid I and II in vitro<br />

and in vivo. Carcinogenesis 9:297-303<br />

Schmutte C, Fishel R (1999) Genomic instability: first step to carcinogenesis. Anticancer Res 19:4665-<br />

4696<br />

Schroeder A, Mueller O, Stocker S, Salowsky R, Leiber M, Gassmann M, Lightfoot S, Menzel W, Granzow<br />

M, Ragg T (2006) The RIN: an RNA integrity number for assigning integrity values to RNA<br />

measurements. BMC Mol Biol 7:3<br />

Schutze K, Posl H, Lahr G (1998) Laser micromanipulation systems as universal tools in cellular and<br />

molecular biology and in medicine. Cell Mol Biol (Noisy-le-grand) 44:735-746<br />

Sebekova K, Wagner Z, Schupp N, Boor P (2007) Genomic damage and malignancy in end-stage renal<br />

failure: do advanced glycation end products contribute? <strong>Kidney</strong> Blood Press Res 30:56-66<br />

Sedgwick B, Bates PA, Paik J, Jacobs SC, Lindahl T (2007) Repair <strong>of</strong> alkylated DNA: recent advances.<br />

DNA Repair (Amst) 6:429-442<br />

Semenza GL (2002) HIF-1 and tumor progression: pathophysiology and therapeutics. Trends Mol Med<br />

8:S62-67<br />

Sen B, Wolf DC, Hester SD (2004) The transcriptional pr<strong>of</strong>ile <strong>of</strong> the kidney in Tsc2 heterozygous mutant<br />

Long Evans (Eker) rats compared to wild-type. Mutat Res 549:213-224<br />

Shah OJ, Wang Z, Hunter T (2004) Inappropriate activation <strong>of</strong> the TSC/Rheb/mTOR/S6K cassette induces<br />

IRS1/2 depletion, insulin resistance, and cell survival deficiencies. Curr Biol 14:1650-1656<br />

196


Chapter 10: References<br />

Shank RC, Magee PN (1967) Similarities between the biochemical actions <strong>of</strong> cycasin and<br />

dimethylnitrosamine. Biochem J 105:521-527<br />

Shibutani S, Takeshita M, Grollman AP (1991) Insertion <strong>of</strong> specific bases during DNA synthesis past the<br />

oxidation-damaged base 8-oxodG. Nature 349:431-434<br />

Sieber SM, Correa P, Dalgard DW, McIntire KR, Adamson RH (1980) Carcinogenicity and hepatotoxicity<br />

<strong>of</strong> cycasin and its aglycone methylazoxymethanol acetate in nonhuman primates. J Natl <strong>Cancer</strong><br />

Inst 65:177-189<br />

Singh SC (1980) Deformed dendrites and reduced spine numbers on ectopic neurones in the<br />

hippocampus <strong>of</strong> rats exposed to methylazoxymethanol-acetate. A Golgi-Cox study. Acta<br />

Neuropathol 49:193-198<br />

Singh SK, Clarke ID, Terasaki M, Bonn VE, Hawkins C, Squire J, Dirks PB (2003) Identification <strong>of</strong> a cancer<br />

stem cell in human brain tumors. <strong>Cancer</strong> Res 63:5821-5828<br />

Sionov RV, Haupt Y (1999) The cellular response to p53: the decision between life and death. Oncogene<br />

18:6145-6157<br />

Sohn OS, Fiala ES, Requeijo SP, Weisburger JH, Gonzalez FJ (2001) Differential effects <strong>of</strong> CYP2E1<br />

status on the metabolic activation <strong>of</strong> the colon carcinogens azoxymethane and<br />

methylazoxymethanol. <strong>Cancer</strong> Res 61:8435-8440<br />

Spatz M (1964) Carcinogenic Effect Of Cycad Meal In Guinea Pigs. Fed Proc 23:1384-1385<br />

Spatz M, Smith DW, McDaniel EG, Laqueur GL (1967) Role <strong>of</strong> intestinal microorganisms in determining<br />

cycasin toxicity. Proc Soc Exp Biol Med 124:691-697<br />

Spiess AN, Mueller N, Ivell R (2003) Amplified RNA degradation in T7-amplification methods results in<br />

biased microarray hybridizations. BMC Genomics 4:44<br />

Steiner G, Suter L, Boess F, Gasser R, de Vera MC, Albertini S, Ruepp S (2004) Discriminating different<br />

classes <strong>of</strong> toxicants by transcript pr<strong>of</strong>iling. Environ Health Perspect 112:1236-1248<br />

Stemmer K, Ellinger-Ziegelbauer H, Ahr HJ, Dietrich DR (2007) Carcinogen-specific gene expression<br />

pr<strong>of</strong>iles in short-term treated Eker and wild-type rats indicative <strong>of</strong> pathways involved in renal<br />

tumorigenesis. <strong>Cancer</strong> Res 67:4052-4068<br />

Stemmer K, Ellinger-Ziegelbauer H, Lotz K, Ahr HJ, Dietrich DR (2006) Establishment <strong>of</strong> a protocol for the<br />

gene expression analysis <strong>of</strong> laser microdissected rat kidney samples with affymetrix genechips.<br />

Toxicol Appl Pharmacol 217:134-142<br />

Stiborova M, Frei E, Arlt VM, Schmeiser HH (2008) Metabolic activation <strong>of</strong> carcinogenic aristolochic acid, a<br />

risk factor for Balkan endemic nephropathy. Mutat Res 658:55-67<br />

Stiborova M, Frei E, Sopko B, Sopkova K, Markova V, Lankova M, Kumstyrova T, Wiessler M, Schmeiser<br />

HH (2003) Human cytosolic enzymes involved in the metabolic activation <strong>of</strong> carcinogenic<br />

aristolochic acid: evidence for reductive activation by human NAD(P)H:quinone oxidoreductase.<br />

Carcinogenesis 24:1695-1703<br />

Stoev SD (1998) The role <strong>of</strong> ochratoxin A as a possible cause <strong>of</strong> Balkan endemic nephropathy and its risk<br />

evaluation. Vet Hum Toxicol 40:352-360<br />

Storkel S, Eble JN, Adlakha K, Amin M, Blute ML, Bostwick DG, Darson M, Delahunt B, Iczkowski K<br />

(1997) Classification <strong>of</strong> renal cell carcinoma: Workgroup No. 1. Union Internationale Contre le<br />

<strong>Cancer</strong> (UICC) and the American Joint Committee on <strong>Cancer</strong> (AJCC). <strong>Cancer</strong> 80:987-989<br />

Studer-Rohr I, Schlatter J, Dietrich DR (2000) Kinetic parameters and intraindividual fluctuations <strong>of</strong><br />

ochratoxin A plasma levels in humans. Arch Toxicol 74:499-510<br />

Sun D, Feng J, Dai C, Sun L, Jin T, Ma J, Wang L (2006) Role <strong>of</strong> peritubular capillary loss and hypoxia in<br />

progressive tubulointerstitial fibrosis in a rat model <strong>of</strong> aristolochic acid nephropathy. Am J Nephrol<br />

26:363-371<br />

Swenberg JA (1993) Alpha 2u-globulin nephropathy: review <strong>of</strong> the cellular and molecular mechanisms<br />

involved and their implications for human risk assessment. Environ Health Perspect 101 Suppl<br />

6:39-44<br />

Swenberg JA, Fryar-Tita E, Jeong YC, Boysen G, Starr T, Walker VE, Albertini RJ (2008) Biomarkers in<br />

toxicology and risk assessment: informing critical dose-response relationships. Chem Res Toxicol<br />

21:253-265<br />

Swenberg JA, Lehman-McKeeman LD (1999) alpha 2-Urinary globulin-associated nephropathy as a<br />

mechanism <strong>of</strong> renal tubule cell carcinogenesis in male rats. IARC Sci Publ:95-118<br />

Szakacs G, Paterson JK, Ludwig JA, Booth-Genthe C, Gottesman MM (2006) Targeting multidrug<br />

resistance in cancer. Nat Rev Drug Discov 5:219-234<br />

Takahashi M, Teh BT, Kanayama HO (2006) Elucidation <strong>of</strong> the molecular signatures <strong>of</strong> renal cell<br />

carcinoma by gene expression pr<strong>of</strong>iling. J Med Invest 53:9-19<br />

Takahashi Y, Okimura Y, Mizuno I, Takahashi T, Kaji H, Uchiyama T, Abe H, Chihara K (1996) Leptin<br />

induces tyrosine phosphorylation <strong>of</strong> cellular proteins including STAT-1 in human renal<br />

adenocarcinoma cells, ACHN. Biochem Biophys Res Commun 228:859-864<br />

Tan MH, Rogers CG, Cooper JT, Ditlev JA, Maatman TJ, Yang X, Furge KA, Teh BT (2004) Gene<br />

expression pr<strong>of</strong>iling <strong>of</strong> renal cell carcinoma. Clin <strong>Cancer</strong> Res 10:6315S-6321S<br />

197


Chapter 10: References<br />

Thomas GV (2006) mTOR and cancer: reason for dancing at the crossroads? Curr Opin Genet Dev 16:78-<br />

84<br />

Todd R, Wong DT (1999) Oncogenes. Anticancer Res 19:4729-4746<br />

Tran H, Brunet A, Griffith EC, Greenberg ME (2003) The many forks in FOXO's road. Sci STKE 2003:RE5<br />

Tsujimura K, Asamoto M, Suzuki S, Hokaiwado N, Ogawa K, Shirai T (2006) Prediction <strong>of</strong> carcinogenic<br />

potential by a toxicogenomic approach using rat hepatoma cells. <strong>Cancer</strong> Sci 97:1002-1010<br />

Turk PW, Laayoun A, Smith SS, Weitzman SA (1995) DNA adduct 8-hydroxyl-2'-deoxyguanosine (8hydroxyguanine)<br />

affects function <strong>of</strong> human DNA methyltransferase. Carcinogenesis 16:1253-<br />

1255<br />

Uchida E, Hirono I (1981) Effect <strong>of</strong> phenobarbital on the development <strong>of</strong> neoplastic lesions in the liver <strong>of</strong><br />

cycasin-treated rats. J <strong>Cancer</strong> Res Clin Oncol 100:231-238<br />

Um SH, Frigerio F, Watanabe M, Picard F, Joaquin M, Sticker M, Fumagalli S, Allegrini PR, Kozma SC,<br />

Auwerx J, Thomas G (2004) Absence <strong>of</strong> S6K1 protects against age- and diet-induced obesity<br />

while enhancing insulin sensitivity. Nature 431:200-205<br />

van Delft JH, van Agen E, van Breda SG, Herwijnen MH, Staal YC, Kleinjans JC (2004) Discrimination <strong>of</strong><br />

genotoxic from non-genotoxic carcinogens by gene expression pr<strong>of</strong>iling. Carcinogenesis 25:1265-<br />

1276<br />

Van Gaal LF, Mertens IL, De Block CE (2006) Mechanisms linking obesity with cardiovascular disease.<br />

Nature 444:875-880<br />

Van Gelder RN, von Zastrow ME, Yool A, Dement WC, Barchas JD, Eberwine JH (1990) Amplified RNA<br />

synthesized from limited quantities <strong>of</strong> heterogeneous cDNA. Proc Natl Acad Sci U S A 87:1663-<br />

1667<br />

Vanherweghem JL, Depierreux M, Tielemans C, Abramowicz D, Dratwa M, Jadoul M, Richard C,<br />

Vandervelde D, Verbeelen D, Vanhaelen-Fastre R, et al. (1993) Rapidly progressive interstitial<br />

renal fibrosis in young women: association with slimming regimen including Chinese herbs.<br />

Lancet 341:387-391<br />

Walker C, Goldsworthy TL, Wolf DC, Everitt J (1992) Predisposition to renal cell carcinoma due to<br />

alteration <strong>of</strong> a cancer susceptibility gene. Science 255:1693-1695<br />

Wang Z, Jiang T, Li J, Proctor G, McManaman JL, Lucia S, Chua S, Levi M (2005) Regulation <strong>of</strong> renal lipid<br />

metabolism, lipid accumulation, and glomerulosclerosis in FVBdb/db mice with type 2 diabetes.<br />

Diabetes 54:2328-2335<br />

Ward JM (2007) The Two-Year Rodent Carcinogenesis Bioassay . Will It Survive? J Toxicol Pathol 20:13-<br />

19<br />

Weisburger EK (1978) Mechanisms <strong>of</strong> chemical carcinogenesis. Annu Rev Pharmacol Toxicol 18:395-415<br />

Whiting MG (1963) Toxicity <strong>of</strong> Cycads. Economic Bot 17:270-329<br />

Williams GM (1999) Chemically induced preneoplastic lesions in rodents as indicators <strong>of</strong> carcinogenic<br />

activity. IARC Sci Publ:185-202<br />

Williams GM, Iatropoulos MJ, Jeffrey AM (1995) Thresholds for DNA-Reactive (Genotoxic) Organic<br />

Carcinogens. J Toxicol Pathol 18:69-77<br />

Williams JN, Jr., Laqueur GL (1965) Response Of Liver Nucleic Acids And Lipids In Rats Fed Cycas<br />

Circinalis L. Endosperm Or Cycasin. Proc Soc Exp Biol Med 118:1-4<br />

Wolf DC, Goldsworthy TL, Donner EM, Harden R, Fitzpatrick B, Everitt JI (1998) Estrogen treatment<br />

enhances hereditary renal tumor development in Eker rats. Carcinogenesis 19:2043-2047<br />

Wolf DC, Goldsworthy TL, Janszen DB, Harden R, Donner EM, David CS, Everitt JI (2000) Promotion by<br />

sodium barbital induces early development but does not increase the multiplicity <strong>of</strong> hereditary<br />

renal tumors in Eker rats. Carcinogenesis 21:1553-1558<br />

Wullschleger S, Loewith R, Hall MN (2006) TOR signaling in growth and metabolism. Cell 124:471-484<br />

Yeung RS, Xiao GH, Everitt JI, Jin F, Walker CL (1995) Allelic loss at the tuberous sclerosis 2 locus in<br />

spontaneous tumors in the Eker rat. Mol Carcinog 14:28-36<br />

Yeung RS, Xiao GH, Jin F, Lee WC, Testa JR, Knudson AG (1994) Predisposition to renal carcinoma in<br />

the Eker rat is determined by germ-line mutation <strong>of</strong> the tuberous sclerosis 2 (TSC2) gene. Proc<br />

Natl Acad Sci U S A 91:11413-11416<br />

Young AN, Master VA, Paner GP, Wang MD, Amin MB (2008) Renal epithelial neoplasms: diagnostic<br />

applications <strong>of</strong> gene expression pr<strong>of</strong>iling. Adv Anat Pathol 15:28-38<br />

Zedeck MS, Brown GB (1977) Methylation <strong>of</strong> intestinal and hepatic DNA in rats treated with<br />

methylazoxymethanol acetate. <strong>Cancer</strong> 40:2580-2583<br />

Zedeck MS, Sternberg SS, Poynter RW, McGowan J (1970) Biochemical and pathological effects <strong>of</strong><br />

methylazoxymethanol acetate, a potent carcinogen. <strong>Cancer</strong> Res 30:801-812<br />

Zhang H, Cifone MA, Murli H, Erexson GL, Mecchi MS, Lawlor TE (2004) Application <strong>of</strong> simplified in vitro<br />

screening tests to detect genotoxicity <strong>of</strong> aristolochic acid. Food Chem Toxicol 42:2021-2028<br />

198


Erklärung<br />

Die vorliegende Arbeit wurde ohne unzulässige Hilfe Dritter und ohne Benutzung anderer<br />

als der angegebenen Hilfsmittel angefertigt. Die aus anderen Quellen direkt oder indirekt<br />

übernommenen Daten und Konzepte sind unter Angabe der Quelle gekennzeichnet.<br />

Weitere Personen, insbesondere Promotionsberater, waren an der inhaltlich materiellen<br />

Erstellung dieser Arbeit nicht beteiligt. Die Arbeit wurde weder im In- noch im Ausland in<br />

gleicher oder ähnlicher Form einer anderen Prüfungsbehörde vorgelegt.<br />

Eigenabgrenzung/ Kooperationen<br />

• Die Hybridisierung der Microarrays (siehe Kapitel 3, 4, 5 und 6) erfolgte in der<br />

Abteilung <strong>Molecular</strong> and Special Toxicology, Bayer Healthcare AG in Wuppertal.<br />

• Sämtliche Paraffinschnitte wurden von Mitarbeitern der Abteilung Non-Clinical Drug<br />

Safety bei Boehringer Ingelheim in Biberach hergestellt.<br />

• LC-MS/MS Analysen zur Detektion von O6MG Addukten (Kapitel 4) wurden von<br />

Mitarbeitern von Pr<strong>of</strong>. Dr. James A. Swenberg am Department <strong>of</strong> Environmental<br />

Sciences and Engineering, University <strong>of</strong> North Carolina, Chapel Hill, USA<br />

durchgeführt.<br />

• Die Exposition der Ratten mit fetthaltiger Diät, sowie die Messung der Blutparameter<br />

(Kapitel 7) wurde von Mitarbeitern von Pr<strong>of</strong>. Dr. Matthias Tschöp, im Department <strong>of</strong><br />

Psychiatry, Obesity Research Centre in Cincinnati, Ohio, USA durchgeführt.<br />

Alle weiteren Leistungen wurden, s<strong>of</strong>ern nicht explizit angemerkt, von mir selbst<br />

durchgeführt.<br />

Konstanz im Oktober 2008<br />

Kerstin Stemmer<br />

199


Danksagung<br />

Meinen herzlichsten Dank möchte ich all denen aussprechen, die mich auf dem Weg zur<br />

Entstehung dieser Doktorarbeit unterstützt haben. Mein besonderer Dank gilt hierbei:<br />

• Pr<strong>of</strong>. Dr. Daniel Dietrich für die Überlassung des Promotionsthemas, seine stete<br />

wissenschaftliche und menschliche Unterstützung, seine Förderung,<br />

Diskussionsbereitschaft und Offenheit gegenüber neuen Projektideen. Insgesamt für<br />

eine schöne und sehr lehrreiche Zeit in der AG-Dietrich, an die ich immer gerne<br />

zurückdenken werde.<br />

• Pr<strong>of</strong>. Dr. Christ<strong>of</strong> Hauck für die Übernahme des Koreferates und sein Interesse an<br />

dieser Arbeit.<br />

• Dr. Dr. Hans-Jürgen Ahr, Dr. Heidrun-Ellinger Ziegelbauer und Mitarbeitern für eine<br />

<strong>of</strong>fene und nette Projektkooperation, ihre stete Diskussionsbereitschaft auch nach<br />

Projektabschluss, das Einlernen in die komplexe Auswertung von Microarray Analysen,<br />

nicht zuletzt für die herzliche Gastfreundschaft und Unterstützung während vieler<br />

mehrwöchiger Aufenhalte bei der Bayer HealthCare AG in Wuppertal.<br />

• Pr<strong>of</strong>. Dr. Matthias Tschöp, Dr. Paul Pfluger und Dr. Diego Perez-Tilve für Ihre<br />

Unterstützung und Begeisterung, ein neues, gemeinsames Projekt auf den Weg zu<br />

bringen, ihre Gastfreundschaft, und die Überlassung eines Laborplatzes während<br />

mehrer Aufenthalte am Obesity Research Center in Cincinnati, sowie für den Einblick in<br />

die Kunst des Cabrewings. Besonders bei Paul möchte ich mich für die Korrekturen und<br />

die konstruktive Kritik an dieser Arbeit bedanken. Nicht zuletzt für unzählige<br />

wissenschaftliche Diskussionen, die so manch gute Idee und Fortschritt mit sich<br />

brachten. Erin Grant möchte ich herzlichst für die Englisch-Korrekturen danken.<br />

• Dr. Ulrich Deschl, Dr. Thomas Nolte und Mitarbeitern, für die Ermöglichung eines 2wöchigen<br />

Arbeitsaufenthaltes in der Abteilung für Non-Clinical Drug Safety bei<br />

Boehringer Ingelheim in Biberach. Ohne diesen waere mir die Planung und<br />

Durchführung der in vivo Studie für diese Doktorarbeit weitaus schwerer gefallen. Nicht<br />

zuletzt bedanke ich mich für das Anfertigen zahlreicher Paraffinschnitte.<br />

• Pr<strong>of</strong>. Dr. James A. Swenberg und Mitarbeitern vom Department <strong>of</strong> Environmental<br />

Sciences and Engineering, University <strong>of</strong> North Carolina, Chapel Hill, danke ich für die<br />

Zusammenarbeit und die Quantifizierung von DNA-Addukten mittels LC-MS/MS.<br />

• Allen ehemaligen und jetzigen Mitarbeiter/-innen der Arbeitsgruppe Dietrich für das<br />

nette und familiäre Arbeitsklima. Besonders Tanja Lampertsdörfer und Gudrun von<br />

Scheven möchte ich für die tatkräftige Unterstützung beim in vivo Versuch und bei der<br />

Verabreichung tausender Schlundsonden danken. Beiden, sowie Dan Dietrich, Alex<br />

Heussner und Evelyn O’Brien gilt mein Dank für die Mithilfe bei den Sektionen.<br />

Und vor allem meiner Familie und meinem Freund Paul für ihre stete Unterstützung in allen<br />

Lebenslagen, ihren stetigen Beistand, und ihre Ruhe während aller Höhen und Tiefen dieser<br />

Doktorarbeit.<br />

200

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!