DISSERTATION

submitted to the Combined Faculties for the Natural Sciences and for Mathematics of the Ruperto-Carola University of Heidelberg, Germany for the degree of Doctor of Natural Sciences

ELUCIDATION OF THE POTENTIAL OF SGK1 AS

PROGNOSTIC MARKER AND THERAPEUTIC

TARGET IN MEDULLOBLASTOMA

Presented by Dipl.-Biol. Sabrina Valesca Pleier Born in Mühlacker, Germany

DISSERTATION

submitted to the Combined Faculties for the Natural Sciences and for Mathematics of the Ruperto-Carola University of Heidelberg, Germany for the degree of Doctor of Natural Sciences

Presented by Dipl.-Biol. Sabrina Valesca Pleier Born in Mühlacker, Germany

Day of oral examination: July 20th, 2012

ELUCIDATION OF THE POTENTIAL OF SGK1 AS

PROGNOSTIC MARKER AND THERAPEUTIC

TARGET IN MEDULLOBLASTOMA

Referees: Prof. Dr. Werner Buselmaier Prof. Dr. Peter Lichter

Declaration

I hereby declare that I have written the submitted dissertation ‘Elucidation of the potential of SGK1 as prognostic marker and therapeutic target in medullo- blastoma’ myself and in this process have used no other sources or materials than those expressly indicated. I hereby declare that I have not applied to be examined at any other institution, nor have I used the dissertation in this or any other form at any other institution as an examination paper, nor submitted it to any other faculty as a dissertation.

______(Place, Date) Sabrina Valesca Pleier

To my family

Contents

TABLE OF CONTENTS

Table of Contents ...... I

Abbreviations ...... IV

Definitions ...... VII

Summary ...... IX

Zusammenfassung ...... X

1 Introduction ...... 1 1.1 Cancer ...... 1 1.1.1 The Hallmarks of Cancer ...... 1 1.1.2 Cellular Oncogenes ...... 6 1.1.3 Tumor Suppressor ...... 7 1.2 Medulloblastoma ...... 9 1.2.1 Histopathology of Medulloblastoma ...... 10 1.2.2 Cellular Origin of Subtypes ...... 12 1.2.3 Molecular Subgroups of Medulloblastoma ...... 16 1.2.4 Treatment of Medulloblastoma ...... 22 1.3 Objective ...... 23 1.3.1 6q Status Defines Medulloblastoma Subgroups ...... 23 1.3.2 The Candidate SGK1 ...... 24 1.3.3 (Patho-) Physiological Role of SGK1 ...... 26 1.4 Aims of the Study ...... 28

2 Materials & Methods ...... 29 2.1 Materials ...... 29 2.1.1 Antibodies ...... 29 2.1.2 Biochemicals and Reagents ...... 29 2.1.3 Buffers and Solutions ...... 31 2.1.4 Cell Culture Reagents ...... 32 2.1.5 Cell Lines and Bacterial Strains ...... 32 2.1.6 Drugs ...... 33 2.1.7 ...... 33 2.1.8 Instruments ...... 33

I

Contents

2.1.9 Materials ...... 34 2.1.10 Molecular Biology Kits ...... 35 2.1.11 Plasmids and shRNAs ...... 35 2.1.12 Primers ...... 36 2.1.13 Software and Databases ...... 36 2.2 Molecular Biology Methods ...... 37 2.2.1 DNA Procedures ...... 37 2.2.2 RNA Procedures ...... 41 2.2.3 Lentivirus-based Procedures ...... 44 2.3 Cell Biology Methods ...... 46 2.3.1 Cell Culture Procedures ...... 46 2.3.2 Functional Analyses ...... 48 2.4 Protein Biochemistry Methods ...... 52 2.4.1 Protein Procedures ...... 52 2.4.2 Immunostaining Prodedure ...... 53 2.5 Statistical Analyses ...... 54

3 Results ...... 55 3.1 Identification of SGK1 as Candidate Gene in Medulloblastoma ...... 55 3.1.1 SGK1 mRNA Expression is correlated with Chromosome 6q ...... Copy-Number Status in Medulloblastoma ...... 55 3.1.2 SGK1 is Differentially Expressed Between Medulloblastoma ...... Molecular Subgroups ...... 56 3.1.3 SGK1 Protein Expression is correlated with ...... Overall Survival of Medulloblastoma Patients ...... 57 3.1.4 SGK1 is Differentially Methylated Across Medulloblastoma ...... Subgroups ...... 58 3.1.5 Medulloblastoma Cell Lines Exhibit Different Variable SGK1 ...... Expression Levels and Inducibility by Treatment ...... 59 3.2 Functional Analyses after Modulating SGK1 ...... in Medulloblatoma Cell Lines ...... 61 3.3 Phenotypic Assays after Stable Overexpression of ...... SGK1 in Medulloblastoma Cell Lines ...... 61 3.3.1 SGK1 Overexpression has no Effect on Proliferation ...... 61 3.3.2 SGK1 Overexpression leads to Increased Migration Rates ...... 62

II

Contents

3.3.3 SGK1 Overexpression has no Effect on Irradiation - ...... but on Chemotherapy Resistance ...... 64 3.4 Lentivirus-mediated Knockdown of SGK1 ...... Induces in Medulloblastoma Cell Lines ...... 67 3.5 Pharmacological Inhibition of SGK1 by ...... a Small Molecule (GSK650394) Reduces Cell Viability in vitro ...... 71 3.6 NDRG1 is a Direct Downstream Target of SGK1 ...... 72 3.7 Expression Profiling: Deregulated Genes After SGK1 Knockdown ...... 73

4 Discussion ...... 78 4.1 SGK1 is a Prognostic Marker for High-Risk-Medulloblastoma ...... 79 4.2 SGK1 is a potential drug target in Medulloblastoma ...... 80 4.2.1 Knockdown of SGK1 Induces Apoptosis in vitro ...... 80 4.2.2 Overexpression of SGK1 Induces Chemotherapy Resistance ...... and Increases Migration Rates in vitro ...... 81 4.2.3 How Could the Obtained Phenotypes be Mediated by SGK1? ...... 82 4.2.4 Small Molecule Inhibitor GSK650394 Induces Apoptosis ...... in Medulloblastoma Cell Lines ...... 85

5 Conclusions and Outlook ...... 87

6 References ...... 88

7 Appendix ...... 100

8 Acknowledgements ...... 105

III

Glossary

ABBREVIATIONS

°C Celsius 4-HC 4-Hydroperoxycyclophosphamide 7-AAD 7-Aminoactinomycin aa Amino acid AcGFP Green fluorescent protein AKT V-akt murine thymoma viral oncogene homolog 1 APC Adenomatous polyposis coli APS Ammoniumperoxodisulfat Array CGH array comparative genomic hybridization ATCC American Type Culture Collection ATM Ataxia telangiectasia mutated ATP Adenosine 5’-triphosphate BAC Bacterial artificial chromosome BAD BCL2-associated agonist of cell death BCA Bicinchoninic acid BCL2 B-cell CLL/lymphoma2 Bp BRCA Breast cancer gene BSA Bovine serum albumin cAMP Adenosine 3’,5’-cyclic monophosphate CB Cerebellum CBP CREB-binding protein CCNU N-(2-chloroethyl)-N'-cyclohexyl-N-nitrosourea, CDKN2A Cyclin-dependent kinase inhibitor 2a cDNA Complementary DNA CMV Cytomegalie-Virus CNS Central nervous system CO2 Carbon dioxide cPPT Central polypurintract CR Conserved region CREB cAMP responsive element-binding protein CREBBP CREB-binding protein CT Cycle of threshold CTNNB1 Beta-catenin Cy Cyanin Cy3/Cy5 Fluorescent dyes of the cyanine dye group Da Dalton DAB Diaminobenzidine DAPI 4´,6-diamidino-2´-phenyllindol-dihydrochloride ddH2O Double distilled water ddNTP 2’,3’-Didesoxyribonukleosid-5’-Triphosphat DEX Dexamethasone

IV

Glossary

DKFZ Deutsches Krebsforschungszentrum DMEM Dulbecco‘s Modified Eagle Medium DMSO Dimethylsulfoxid DNA Deoxyribonucleic acid dNTP 2’-Desoxyribonukleosid-5’-Triphosphat ds Double stranded DTT Dithioreitol E. coli Escherichia coli EC50 Half maximal effective concentration ECL Electrochemiluminescence EDTA Ethylendiaminetetraacetic acid EGFR Epidermal growth factor receptor EGL External granule layer env Envelope ErbB2 Human epidermal growth factor receptor 2 ERK Extracellular signal-regulated kinase ERα Estrogen receptor alpha et al. et alteres EtOH Ethanol FACS Fluorescence-activated cell sorting FCS Fetal calf serum FSC Forward scatter fwd Forward g Gramm GC Glucocorticoid GFP green fluorescent protein GLI2 GLI family zinc finger 2 GR h hours H2O Water HA Viral hemagglutinin protein HE Hematoxylin and eosin HRP Horse radish peroxidase Ig Immunoglobulin IHC Immunohistochemistry kb Kilo base pairs (103bp) kDa Kilodalton l Liter LB Luria-Bertani LFS Li-Fraumeni Syndrome LTR Long terminal repeat m Milli M Molar (mol/l) mA Milliampere MAPK Mitogen activated protein kinase MB Medulloblastoma

V

Glossary

mcs Multiple cloning site MEK Mitogen-activated protein kinase kinase min Minutes ml Milliliter MOI Multiplicity of infection mRNA Messeger RNA MYC V-myc myelocytomatosis viral oncogene homolog n/a Not applicable NaCl Sodium chloride NCBI National Center for Biotechnology NDRG1 N-myc downstream regulated 1 NFkB Nuclear factor of kappa light polypeptide gene enhancer in B-cells nm Nanometer O2 Molecular oxygen OD Optical density ORF Open reading frame OS Overall survival p Short arm of a chromosome PAA Polyacrylamide PARP Poly(ADP-ribose) polymerase1 PBS Phosphate buffered Saline PCR Polymerase Chain Reaction PDGF Plateled-derived growth factor receptor PDK Pyruvate dehydrogenase kinase PE Phycoerythrin PFA Paraformaldehyd PFS Progression free survival PI Phosphatidylinositol PI Propidiumiodid Pi3K Phosphoinositide-3-kinase PIP Phosphatidylinositol phosphate PMSF Phenylmethylsulfonylfluorid pol Polymerase PTCH Patched PTEN Phosphatase tensin homolog PVDF polyvinylidene fluoride q Long arm of a chromosome qRT-PCR Quantitative real time PCR RAS Rat sarcoma viral oncogene homolog rev Reverse RNA Ribonucleic acid RIN RNA Integrity Number RNAi RNA interference Rnase Ribonuclease rpm Rounds per minute RSV Rous Sarcoma Virus

VI

Glossary

RT Reverse transcription RT Room temperature RTK Receptor Tyrosine Kinase SDS Sodium dodecyl sulfate SDS-PAGE Sodium dodecyl sulphate polyacrylamide gel electrophoresis sec Seconds SFRP1 Secreted frizzled-related protein 1 SGK1 Serum/glucocorticoid regulated kinase 1 SGZ Subgranular zone SHH Sonic hedgehog shRNA Short hairpin RNA SOC Super optimal broth with catabolite SSC Sideward scatter SUFU Suppressor of fused homolog SVZ Suventricular zone TBE TRIS-Borat-EDTA TBE Tris base - boric acid - EDTA TBS Ttris buffered saline TBS-T Tris buffered saline plus Tween-20 TE Tris - EDTA TEMED N,N,N,N- tetramethylethylendiamine TP53 Tumor protein TRC The RNAi Consortium Tris Tris-(hydroxymethyl)-aminomethan U Unit UV Ultraviolet V Volt v-myc Myelocytomatosis viral oncogene homolog v-src Avian sarcoma viral oncogene v/v Volume per volume VEGF Vascular endothelial growth factor w/v Weight per volume WHO World Health Organization WNT Wingless-type MMTV (mouse mammary tumor μ Mikro

DEFINITIONS

Gene symbols written as upper-case letters indicate human protein (e.g. SGK1). Gene symbols written as italicized upper-case letters indicate human gene (e.g. SGK1). Gene symbols written capitalized indicate murine protein (e.g. Sgk1). Gene symbols written capitalized and italicized indicate mouse gene (e.g.Sgk1).

VII

Glossary

VIII

Summary

SUMMARY

Medulloblastoma comprises the most common malignant brain tumor in children and one of the leading causes of cancer-related mortality in this age group. Despite significant therapeutic achievements within the last three decades, medulloblastoma is still associated with a comparably poor overall survival rate of only 70%. Moreover, many patients suffer from severe adverse effects from adjuvant chemo- or radiotherapy. A better understanding of the underlying molecular pathomechanisms could reveal more targeted therapeutic approaches for molecular subgroups to enhance individual outcome. In the present study, comparative array-CGH and genome-wide expression profiling analyses (n=64) demonstrated that copy-number aberrations at the genomic locus of serum- and glucocorticoid regulated kinase (SGK1) at 6q23 were tightly correlated with mRNA abundance. Immunohistochemical examinations in an indepen- dent patient cohort (n=260) and correlation with follow-up data revealed a significant association of SGK1 immunopositivity with poor overall survival as assessed by Kaplan- Meier analysis (p = 0.0168). Based on this integrative genomics approach, SGK1 was suggested as a novel prognostic marker, and SGK1 immunohistochemistry may serve as a powerful diagnostic tool in medulloblastoma patients. SGK1 encodes a kinase with high sequence- and structural homology to the anti-apoptotic kinase AKT/PKB and, as the name implicates, it is transcriptionally up-regulated by serum and . SGK1 was previously found to play a role in promoting cell survival and cell-cycle progression by phosphorylating diverse downstream targets such as FOXO3a, GSK-3β, p27, and NDRG1. Functional analyses in vitro conducted in this project demonstrated that loss of SGK1 function causes apoptosis induction and that stable overexpression of SGK1 promotes migration. Furthermore, a presumptive impairment in chemotherapy sensitivity, caused by ectopic SGK1 overexpression, has been observed in medullo- blastoma cells. These findings provide in vitro evidence for a crucial role of SGK1 in medulloblastoma biology making it a prime candidate for targeted therapy in high-risk medulloblastoma patients.

IX

Zusammenfassung

ZUSAMMENFASSUNG

Medulloblastome sind die häufigsten malignen Hirntumoren, die im Kindesalter auftreten und stehen an der Spitze der krebsassoziierten Todesursachen dieser Altersgruppe. Trotz bedeutender therapeutischer Errungenschaften während der letzten drei Jahrzehnte, sind Medulloblastome immernoch mit verhältnismäßig schlechten Gesamtüberlebensraten von nur 70% verbunden. Darüber hinaus kommt es bei vielen Patienten zu starken Nebenwirkungen von Radio- und Chemotherapie. Ein besseres Verständnis der zugrundeliegenden molekularen Pathomechanismen könnte spezi- fischere zielgerichtete Therapieansätze für bestimmte Subgruppen aufzeigen, um einzelne Befunde zu verbessern. In der vorliegenden Studie haben vergleichende Analysen von genomischen Aberrationen mit korrespondierenden Expressionsprofilen (n=64) ergeben, dass der DNA-Kopienzugewinn im Genlocus von Serum- und Gluko- kortikoid-regulierter Proteinkinase (SGK1) auf Chromosom 6q23 mit mRNA Abundanz korreliert ist. Immunohistochemische Untersuchungen in einer unabhängigen Patienten- kohorte (n=260) wurden mit klinischen Daten korreliert und ergaben eine signifikante Assoziation von SGK1 Immunfärbung mit schlechter Überlebensrate, die mit Hilfe der Kaplan-Meier Analyse bestimmt wurde (p = 0.0168). Basierend auf diesem integrativen Ansatz konnte SGK1 als neuer prognostischer Biomarker vorgeschlagen werden. Dabei könnte die SGK1 Immunfärbung als vielversprechendes diagnostisches Hilfsmittel bei Medulloblastompatienten dienen. SGK1 codiert für eine Kinase mit hoher Sequenz- und Strukturhomologie zur anti-apoptotischen Kinase AKT/PKB. Wie der Name bereits impliziert, wird sie auf transkriptioneller Ebene durch Serum und Glukokortikoide hochreguliert. SGK1 wurde sowohl eine Pro-Survival Rolle, als auch positive Be- einflussung der Zellzyklus-Progression zugeschrieben. Diese Effekte werden durch Phosphorylierung verschiedener Zielmoleküle, wie zum Beispiel FOXO3a, GSK-3β, p27 und NDRG1, vermittelt. Funktionelle in vitro Analysen in dieser Studie haben gezeigt, dass der Verlust von SGK1-Funktionalität Apoptose induziert und SGK1 Überexpression die Migration fördert. Des Weiteren wurde eine mutmaßliche Beeinträchtigung der Sensitivität gegenüber Chemotherapeutika festgestellt, die durch ektopische Über- expression von SGK1 in Medulloblastom Zelllinien hervorgerufen wurde. Diese Erkennt- nisse liefern in vitro Beweise dafür, dass SGK1 eine entscheidende Rolle in der Biologie des Medulloblastoms spielt. Deshalb ist es ein wichtiger Kandidat für zielgerichtete Therapie in Hochrisiko-Medulloblastompatienten.

X

Introduction

1 INTRODUCTION

1.1 CANCER

Multicellular organisms hold complex mechanisms allowing proper development, survival and maintenance of cellular homeostatis. There is a manifold interplay of diverse cells within the body, which needs to be tightly regulated because failures can have harmful consequences such as cancer. Malignant cells generally constitute a neoplastic burden, which can develop within almost every tissue in the body. To date, more than 200 different known human cancer entities have been described1. The majority of human tumors derives from epithelial tissues and is named carcinomas. Other solid tumors arising from the mesenchyme are referred to as sarcomas. Another group of non-epithelial cancers comprises tumors of the nervous system, which consist of ectodermally derived tissue. These entities are called according to their proposed cell of origin. Thus, medulloblastoma refers to the medulloblast, a progenitor cell that is proposed to build the medulla oblongata. Furthermore, malignancies can occur from the hematopoietic system such as leukemias and lymphomas2.

1.1.1 The Hallmarks of Cancer Tumorigenesis is a multistep process with each step being an advancement for a normal cell on the way to turn into a malignant progeny. Thus, for the transformation into a malignant tumor cell, a microevolution has to be undergone, which is accompanied by the acquisition of several properties enabling unrestricted growth within the host body. Certain features allow tumor cells to evade control mechanisms. Solid tumors commonly exhibit the following characteristics, which have been summarized as ‘hallmarks of cancer’ by Hanahan and Weinberg in 20003 (Figure 1).

1

Introduction

Figure 1 The Hallmarks of Cancer. The illustrated characteristics can be considered as the classical hallmark capabilities of cancer described by Hanahan and Weinberg in 2000. Adapted from Hanahan et al.4

Independence of external growth signals At early oncogenic stages, a dividing cell clone becomes independent of mitogenic growth signals provided by its microenvironment through the acquisition of genetic or epi- genetic alterations. The achievement of self-sufficient proliferation is a key step of tumor initiation and is often referred to as neoplastic transformation. This can be achieved by an autocrine production of growth factors by the cancer cells themselves. Alternative mechanisms are the overexpression or hyperresponsiveness of cell surface receptors or switching on downstream signaling pathways independent of prior growth factor binding to these receptors. For example, the RAS (rat sarcoma) signaling pathway mediates survival and proliferation. It is downstream of various receptor tyrosine kinases and RAS mutations have been found in many human cancers5.

Insensitivity to anti-growth signals The growth advantage of certain sub-clones occurs through further (epi)genetic changes to generate new phenotypes. Another hallmark of cancer cells dealing with signals provided by the microenvironment consists of the circumvention of growth suppressors. In normal tissues, cellular quiescence is maintained by soluble anti-growth factors like TGFβ (transforming growth factor-β) in addition to other extracellular matrix components.

2

Introduction

TGFβ mediates cell cycle arrest in G1-phase to block proliferation, furthermore it induces cell differentiation and apoptosis. Many tumor cells lack anti-growth inhibition by TGFβ caused by mutations, which leads to uncontrolled proliferation6.

Evasion of cell death Beyond autonomous proliferation, tumor cells need to acquire additional capabilities to overcome homeostatic barriers e.g. to circumvent cell death - mainly apoptosis. Apoptosis is the process of programmed cell death and can be triggered by both intrinsic or extrinsic signals. Eventually, cell death procedes in defined steps like membrane blebbing, chromosome degradation and nucleus fragmentation. The accumulation of several abnormalities causes apoptotic crisis in healthy cells, but not in cancer cells - highlighting the evasion of apoptosis as one major hallmark of tumorigenesis7. In normal cells, p53 is a cellular gatekeeper by controling cell cycle progression and apoptosis8. Malignant cells often lack functional p53, thus apoptosis is prevented. Besides evading apoptosis, tumor cells need to avoid other processes such as senescence and autophagy. The latter one is a catabolic mechanism to reallocate nutrients in starving cells9. Senescence is a post-mitotic state, in which the cell has lost its proliferative capacity10.

Unlimited replicative potential Further microevolutionary steps have to be fulfilled in order to build a tumor bulk. Normal differentiated cells can undergo only a defined number of divisions. This is due to the shortening of the telomeres in each DNA replication step. Upon reaching a critical length, replicative senescence is induced. In order to achieve the capability of limitless selfreplication, malignant tumor cells have to adapt in maintaining telomeres for acquiring immortality. To this end, the majority of tumor cells re-activates telomerase, a ribo- nucleoprotein complex that synthesizes telomeric DNA. Another mechanism is the recombination-based process of alternative lengthening of telomeres (ALT)11.

Induction of angiogenesis When a solid tumor has accumulated the previous characteristics and reached a certain dimension, the supply with oxygen and nutrients becomes insufficient. To overcome this deficiency, the tumor undergoes an angiogenic switch from the avascular to the vascular phase. For this, a neo-vascularization process penetrating the tumor mass is needed12. Angiogenesis is triggered and coordinated by a multitude of soluble factors like HIF

3

Introduction

(hypoxia inducible factor), VEGF (vascular endothelial growth factor) or ANG1 (angiopoetin-1), which are often deregulated in tumor cells13.

Tissue invasion and metastasis Another important hallmark of tumor cells is their capability to invade and metastasize14. This ability is only achieved by a small sub-population within the tumor15. Physical boundaries have to be overcome to spread and colonize at distant organs. The first step in metastasis is becoming invasive, which is facilitated by the loss of cell-cell adhesion molecules. The epithelial to mesenchymal transition (EMT) process, in which epithelial cells switch to a mesenchymal progenitor phenotype, empowers malignant cells to detach and enter blood vessels. Furthermore, matrix metalloproteinases (MMPs) play a crucial role in metastasis. Their upregulation enables the cells to overcome the extracellular matrix boundary16. Accordingly, some of the disseminated cancer cells will find a new microenvironment, where they are able to settle down and grow.

Recently, a complementing review to the publication of 2000 has been published by Hanahan and Weinberg. They extend the number of classical cancer hallmarks to include four additional characteristics that enable progression of a neoplasia4 (Figure 2).

Figure 2 Emerging Hallmarks and Enabling Characteristics. Recent research has highlighted the importance of complementary capabilities of cancer additionally to the classical hallmarks. Adapted from Hanahan et al.4

4

Introduction

Immune escape Interaction of premalignant clones with the microenvironment, especially immune cells interfere with tumorigenesis. According to the theory of immune surveillance, the counteracts uncontrolled by secreting ligands, such as members of the TNF (tumor necrosis factor) family17. Every cell is monitored by an alert system of the immune system to erase transformed cells. Consequently, malignant tumor cells need the capability to evade the immune response. Hence, this is another hallmark of cancer.

Deregulation of energy metabolism The ability of cancer cells to switch their energy metabolism from normal aerobic mitochondrial respiration to glycolysis has been found in various tumors. In normal cells, this process only occur under anaerobic conditions, whereas it is a widespread phenomenon in fast-growing cancers even under aerobic conditions. This effect was first described by Otto Warburg in 192418. In the last years, deeper scientific insights reinforced the concept of metabolic reorientation in tumor cells. Although less ATP molecules are produced by glycolysis, there is a beneficial effect in generating diverse biomolecules like amino acids and nucleotides that are necessary for cell proliferation19. In addition, tumor cells counteract the lower ATP production in upregulating glucose transporters, for example GLUT1 (glucose transporter 1), which increases the glucose uptake20,21.

Genome instability Another hallmark that is often found in cancer cells is genomic instability. The defects in genome maintenance and repair accumulating during cancer development provide advantages for the tumor and can support its progression. One reason for genetic instability is the loss of telomeric DNA22. A second explanation is the increased mutation rate in malignant cells and furthermore the lack of a proper cell control system, which normally eliminates defective cells23,24.

Tumor-promoting The immune system also could play a supportive role for the tumor, basically in mediating chronic inflammation. Thereby, it contributes to various capabilities of malig- nant cells such as supplying growth factors, survival factors, angiogenic factors, extracellular matrix modifying enzymes25. This leads in turn to the activation of the above mentioned hallmarks of cancer.

5

Introduction

Notably, not all of the conditions described above have to be fulfilled for the development of a neoplasia. In general, a tumor, which grows within the natural boundaries of its tissue and does not infiltrate into the surrounding, is regarded as benign. Conversely, an invasive tumor phenotype, which is not locally restricted, is referred to as malignant. Different grades of malignancies occur within one type of cancer ranging from benign lesions to highly malignant and infiltrating tumors with the ability to metastasize. The most devastating stage is when a tumor starts to metastasize. This means that cells disseminate to form cancer colonies at distant sites of the body2. The model that the acquisition of fatal alterations occurs in a multistep process was initially demonstrated for colorectal cancer. Benign adenomas can transform to malignant carcinomas by accumulating several mutations over several years26,27. The probability to accumulate various alterations increases with the age of an organism and its exposure to carcino- genic factors. Childhood malignancies exhibit a very early onset of disease, which is therefore an exeptional case. During the development from a healthy towards a malignant cell two classes of genes - tumor suppressor genes and oncogenes - play a critical role in gaining the previously described hallmarks. It is important to distinguish the 'driver' mutations that push cells towards cancer from the 'passenger' mutations that are a by-product of cancer cell development28.

1.1.2 Cellular Oncogenes Under physiological conditions is maintained by balancing cell growth and cell death. A premalignant cell has to overcome a series of barriers to reach sustained and unrestricted cell growth, which gives rise to tumorigenesis. In this process, onco- genes play a crucial role. Per definition oncogenes are cellular genes capable of accelerating proliferation in a dominant way on a cellular level upon activation. Their normal counterparts are referred to as proto-oncogenes. Major oncogenic mechanisms lead to a gain-of-function. This can be achieved by activating genetic mutations, translocations or amplifications resulting in overexpression. Proto-oncogenes are typically implicated in signal transduction promoting cell growth, cell proliferation, survival or induction of developmental processes and encode for proteins of different classes: growth factors, growth factor receptors, signal transducers, transcription factors, chromatin remodelers, and apoptosis regulators29. The first oncogene was discovered in 1970 in a chicken retrovirus, the Rous Sarcoma Virus (RSV), and termed as v-src (avian sarcoma viral oncogene) due to its capability of transforming neoplastic disease in

6

Introduction

chicken when overexpressed30,31. Later studies revealed that in untransformed host cells, a related gene (c-SRC) is present, which originally has been acquired by the virus32,33. The sarcoma viral oncogene homolog RAS is a downstream key mediator of extrinsic growth stimuli. Proteins of the RAS-family conduct mitogenic signals from growth factor receptors and other transmembrane proteins like integrins towards the cytoplasmic cascade of mitogen-activated protein kinases (MAPK)27. The activation of RAS due to aciquisitation of mutations has been described in various cancer types34,35. From a therapeutic point of view, the gain-of-function character of oncogenes offers promising implications in approaching inhibition of their function. Therefore, molecules have been developed to specifically target these proteins allowing rational treatment in contrast to conventional therapies, which are generally aimed at all dividing cells in the patient. This is achieved for example by imatinib/glivec, an ABL kinase inhibitor in CML, gefitinib/iressa targeting the EGF receptor in non small cell lung cancer (NSCLC) or Lapatinib/Tyverb used in breast cancer inhibiting ERBB2. These drugs are already in clinical use but require oncogene addiction of the tumor to this function36.

1.1.3 Tumor Suppressor Genes In contrast to the growth promoting oncogenes, tumor suppressor genes protect a cell from one step on the path to cancer by exhibiting a suppressive function on cellular growth or inducing apoptosis. Therefore, under normal physiological conditions, these genes counteract the growth inducing effects of (proto-) oncogenes2. In tumorigenic pro- cesses, mutations in tumor suppressor genes cause a loss-of-function and the cell can progress to cancer, usually in combination with other genetic changes. Mutant tumor suppressor alleles are usually recessive, whereas mutant oncogene alleles are typically dominant. Consequently, if only one allele for the tumor suppressor gene is damaged, the second one can still give rise to the correct protein. Thus, generally, both alleles of a tumor suppressor gene are inactivated or lost during oncogenesis, which is implied in the two-hit hypothesis introduced 1971 by Alfred Knudson. This discovery was initially found in cases of retinoblastoma caused by biallelic inactivation in the RB gene. Whereas the mutation of one allele is inherited via germ line, the second hit is acquired by spontaneous mutation of the remaining allele37. Nowadays, it is known that RB is a central player in many cancer-relevant pathways such as cell cycle regulation and apoptosis. Disrupted pRb/E2F interaction abrogates cell cycle control38,39. Furthermore, RB functions as an adaptor protein for chromatin remodeling enzymes. The importance

7

Introduction

of RB is reflected by its frequent loss-of-function in many other human cancers besides in retinoblastoma40. Another example is given by the protein PTEN, which is encoded by the tumor suppresssor gene phosphatase and tensin homolog (PTEN). It acts by opposing the action of PI3K, which is essential for anti-apoptotic and pro-tumorigenic AKT activation41. However, there are also exceptions to the two-hit rule for tumor suppressors. A prominent example for this is protein 53 (p53). The TP53 gene product p53 is a short-lived transcription factor, which induces cell cycle arrest, senescence, DNA repair or apoptosis in response to cellular stresses like irradiation, hypoxia or DNA damage. Mutation of only one TP53 allele may lead to loss-of-function in a dominant negative manner because the protein exerts regulation as a homotetramer and this function is already lost by incorporation of already one mutant monomer42,43. If the p53 pathway becomes activated in order to control DNA integrity and cell cycle progression, the protein is stabilized by uncoupling from its negative regulators MDM2 or MDM4, which leads to the accumulation of p53. Finally, homotetramers are formed to act as transcription factor inducing target . Loss of functional p53 is found in more than 50% of all human cancers illustrating that cellular growth control is an important aspect in cancer prevention44,45. Further exceptions to Knudson’s two-hit concept are given by the phenomenon of exhibiting haploinsufficiency such as e.g. PTCH in medulloblastoma and in the p27Kip1 mouse models. Here, already loss of only one allele of the tumor suppressor leads to predisposition for tumors46,47. Hence, the assumption that null mutations in tumor suppressor genes are recessive excludes those genes that exhibit haploinsufficiency. In the vast majority of sporadic cancers, somatic mutations or deletions are found, which occured spontaneously. In contrast, hereditary cancers are caused by germline mutations. Many hereditary cancer syndromes that are characterized by an increased risk of developing cancer have been described: Familial adenomatous polyposis (APC mutation), Li-Fraumeni-Syndrome (TP53 mutation), and Retinoblastoma (RB mutation)48.

Targeting this group of genes therapeutically is much more demanding compared to the pharmacological inhibition of oncogenes, because the function of the gene (product) has to be restored. This could be accomplished by introducing tumor suppressor genes via gene therapy. Another indirect approach is the artificial induction of apoptosis by administration of agents targeting complementary processes as exemplified by the use of poly (ADP-ribose) polymerase (PARP) inhibitors such as olaparib and iniparib in BRCA-mutated breast cancers or PTEN-defective tumors (e.g. prostate cancer)49.

8

Introduction

1.2 MEDULLOBLASTOMA

Medulloblastoma is the most common malignant brain tumor in children. The small blue cell malignancy constitutes a major cause of mortality in pediatric oncology. While rapidly fatal if untreated, current treatment regimens including surgery, state-of-the-art radiation and chemotherapy, result in overall survival rates exceeding 70%50. About 60% of reported cases in the United States (U.S.) occur in patients in the first 14 years of life. Peak age of incidence is 3-5 years and boys are almost twice as likely to develop medulloblastoma as girls. Medulloblastoma in adults is rare, comprising less than 1% of CNS malignancies51. According to the WHO (World Health Organization) classification of tumors of the central nervous system (CNS), medulloblastoma is a grade IV tumor of embryonal origin arising in the cerebellum. It is an invasive and rapidly growing tumor with the tendency to dissiminate through the cerebrospinal fluid (CSF) and metastasizes to other parts of the brain and the spine though this varies between different subtypes. Medulloblastoma growth penetrates the brainstem and the 4th ventricle because of its localization in the posterior fossa52. This brain area controls posture and complex motor functions such as balance and speech. For infants and older children with medulloblastoma, symptoms like lethargy, vomiting and headaches are very likely to occur, as well as problems in walking, speech, writing and double vision due to the tumor. Tumors located in the cerebellum, such as medulloblastoma are also referred to as infratentorial tumors, meaning that they are located below the tentorium. This thick membrane separates the two cerebral hemispheres of the brain from the cerebellum (Figure 3).

9

Introduction

Figure 3 Diagram showing where the tentorium is located in the brain. Modified from CancerHelp UK1.

1.2.1 Histopathology of Medulloblastoma Varying histopathological subtypes seen on biopsies are currently categorized into five main variants of medulloblastoma according to the WHO classification scheme: classic medulloblastoma, desmoplastic medulloblastoma, medulloblastoma with extensive nodularity (MBEN), large cell medulloblastoma, and anaplastic medulloblastoma. Morphologically, classic medulloblastoma is composed of densely packed cells with round-to-oval or carrot-shaped hyperchromatic nuclei marginally surrounded by cytoplasm. Neuroblastic (Homer Wright) rosettes consisting of tumor cell nuclei arranged in a parallel or circular manner are observed in less than 40% of these cases and are often associated with high mitotic activity. Classic medulloblastomas develop in the cerebellar vermis and affect mostly older children. The desmoplastic variant is characterized by focal appearance of nodules with few differentiated neurocytic cells including reticulin-free zones (pale islands) surrounded by internodular regions of densely packed, less differentiated and highly proliferative cells. The nodules represent areas of neuronal maturation exhibiting a reduced nuclear-to-cytoplasmic ratio. The maturated cells show negligible mitotic activity and increased apoptosis. In contrast to classic medulloblastomas, the desmoplastic subtype is mainly found in the hemispheres of the cerebellum and accounts for more than 50% of all cases occurring in infants < 3 years of age. This histologic variation also encompasses the MBEN subtype that was

10

Introduction

previously designated ‘cerebellar neuroblastoma. It is marked by a lobular archticture with numerous and particularly enlarged reticulin-free zones and rich neuropil-like tissue. These nodules of neurocytic cells exhibit a streaming pattern in the fibrillary background, whereas in some areas the internodular component is markedly reduced. The term large cell medulloblastoma describes already the distinctive phenotype of monomorphic cells with large, round, and vesicular nuclei harboring prominent nucleoli. Due to a lack of cohesiveness, the cells of this variant show the highest mitotic and apoptotic rates among all medulloblastoma histotypes. There is a considerable cytologic overlap in features like pleomorphism and high cell turnover to the anaplastic subtype, for which a combined large cell/ anaplastic (LCA) medulloblastoma category has been proposed. The anaplastic variant also includes tumors with atypical cell forms. However, both variations are characterized by an aggressive behavior resulting in poor clinical outcome. The heterogeneity within this group is reflected in the histopathological picture as well as in the variability in the age of onset ranging from below 3 to 16 years of age50. Medullo- myoblastoma (with myogenic differentiation) and melanocytic medulloblastoma (with melanocytic differentiation) complete the histologic spectrum, whereas they are no longer considered separate variants because their phenotypic tumor cells can occur in different other medulloblastoma subtypes and they share genetic profiles. To sum up, there are currently three (since two are combined) main medulloblastoma subtypes distinguished in neuropathology accompanied by well-defined differences in age distribution (Table 1), to which particular ICD-O codes (International Classification of Disease for Oncology)50 were assigned53,54.

Table 1 Histological variants of medulloblastoma and frequencies in different age groups.

Medulloblastoma ICD-O All childhood Children Children 3-16 Median subtype code medulloblastoma < 3 years years age at presentation

Classic 9470/3 73% 26% 78% 8.1 years

Desmoplastic/ 9471/3 10% 57% 5% 2.7 years MBEN

Large cell/ 9474/3 17% 17% 17% 6.8 years anaplastic

11

Introduction

1.2.2 Cellular Origin of Subtypes In the cancer stem cell (CSC) hypothesis context, it is currently thought that embryonal tumors such as medulloblastoma arise from transformed neural stem cells and/or granule neuron progenitors of cerebellar origin, which were prevented from differentiating into their normal physiological cell stage55. In many ways, medulloblastoma and neural stem cells share cellular and molecular similarities. Recent studies show evidence that genetic anomalies in developmental pathways required for the normal maturation of the cerebellar cortex, in particular developmental pathways for granule cell progenitor (GCP) , give rise to medulloblastoma formation. Analysis of murine transgenic lines have confirmed that lineage-restricted progenitors or stem cells of the external granular layer (EGL) can be transformed into a medulloblastoma, via activated sonic hedgehog (SHH) signaling pathway56. Tumors associated with desmoplastic histology originate from GCPs in the EGL that often carry mutations in SHH signaling pathway effectors (PTCH1, Smo). Furthermore, stem and/or progenitor cell populations outside the EGL are also believed to serve as cells of origin for a subset of medulloblastomas. Indeed, many non-nodular tumors express markers suggesting that they are derived from the ventricular zone (VZ) germinal layer, showing evidence of Wnt pathway activation driven by activating mutations in APC/ß-catenin, which are present in a fraction of classic medulloblastomas. Focal high-level amplifications of MYC(N) are frequently associated with LCA medulloblastoma subtype57-60. The ‘medulloblast’ for other subsets of medulloblastoma has yet to be elucidated. An overview of the histopathological medulloblastoma subtypes and their proposed cellular origin is shown in Figure 4. Early postnatal GCPs are a heterogenous population representing a subset of progenitors potentially with different patterns of connectivity and different susceptibility to tumorigenesis. The precise identification of subpopulations of normal GCPs responsible for each subset of medulloblastoma requires genomic clustering studies and massive sequencing analysis of single cells to define genetic differences and alterations61.

12

Introduction

Figure 4 Histopathological subtypes of medulloblastoma and their proposed cellular origin. Stem and/or progenitor cells derived from the ventricular zone (VZ) are thought to give rise mainly to the classic medulloblastoma variant one fraction of these being dependent on APC/ß-catenin mutations activating aberrant WNT signaling. Copy-number changes of chromosome17q/17p and MYC/MYCN amplifications are observed in the classic subtype but may also give rise to LCA medulloblastoma. In contrast, progenitor cells in the external granular layer (EGL) are proposed to give rise to mainly nodular/desmoplastic medullo- blastoma. They achieve malignancy via constitutive sonic hedgehog (SHH) activation. Further acquisition of MYCN amplifications could transform them to LCA tumors. Other cerebellar stem cells or progenitors are discussed to be origin of other medulloblastoma variants (modified from Fan et al. 62 and Louis et al.50

Etiology and Deregulated Pathways in Medulloblastoma Subgroups Although the cause of medulloblastoma is still unclear, several research groups across the globe are making significant progress in understanding its biology. Various genomic aberrations have been identified to play a role in the development of medulloblastoma. For example, the loss of the distal part of chromosome 17, distal to the TP53 gene, possibly accounts for oncogenic transformation of undifferentiated cerebellar cells. Moreover, there are a few rare hereditary tumor syndromes associated with increased risk for developing these tumors. Medulloblastomas were seen in patients with Gorlin syndrome (OMIM #109400), Turcot syndrome (OMIM #276300), as well as Li-Fraumeni syndrome (OMIM #151623)61,63. Table 2 shows an overview of dysfunctional signaling pathways in association with medulloblastoma.

13

Introduction

Table 2 Signaling pathways in cerebellar development and associated dysfunction in medullo- blastoma subtypes. Modified from Hatten et al.61

Signaling Normal pathway Disease Mutations and/or Medullo- Mechanism of pathway function abnormalities blastoma oncogenesis subtype

Wnt Cell proliferation Turcot ß-Catenin, APC WNT Upregulated Syndrome and Monosomy 6 transcription of and MB proliferative genes

SHH/Ptch Cell proliferation Gorlin Germline loss of SHH Increased cell division Syndrome function in Ptch1 and loss of cell cycle and Sufu exit

Notch 2 Cell proliferation Medullo- Notch2, Jag1 Unknown Loss of cell cycle blastoma and Hes1 arrest overexpression

BMP 2, 4 Cell cycle arrest Gorlin BMP pathway SHH Downregulation of and neuronal Syndrome genes expression, loss of differentiation downregulated cell cycle arrest and differentiation

P53 cell cycle control Li- Loss-of-function SHH Failure of cell cycle and DNA damage Fraumeni mutation in TP53 arrest, senescence or response syndrome apoptosis

The first indication that inherited mutations in developmental genes contribute to medulloblastoma was the finding that germ line mutations in the PTCH1 gene pre- dispose Gorlin syndrome patients to develop medulloblastoma. Gorlin syndrome is an inherited condition associated with nevoid basal cell carcinoma in combination with other symptoms and/or malignancies such as medulloblastoma64,65. Focused attention on abnormalities in SHH signaling in hereditary medulloblastoma discovered inactivating germ line mutations in genes coding for the SHH receptor PTCH1, in the G protein- coupled Smoothened receptor (SMO), or occasionally in the SHH inhibitor SUFU66-69. These mutations result in aberrant activation of the SHH pathway. This pathway is crucial for normal formation of the cerebellum, because it governs the proliferation of the most abundant neuronal population in the brain: the granule neuron precursor (GNP) cells. GNPs have been suggested to be the origin of medulloblastoma, arising as an abnormality of the normal development process, due to an aberrant proliferative stimulus created by constituviely active SHH signaling. If a GNP cell remains in the EGL and fails to exit the cell cycle, eventually this event may result in tumorigenesis70. In its normal physiological role, the trans-membrane protein PTCH1 functions as a receptor for SHH. In absence of the SHH ligand, PTCH1 maintains the SHH pathway in the off state by

14

Introduction

acting on smoothened (SMO). Upon SHH binding to the receptor, this repression is released and a signal is transduced through SMO to the nucleus thereby increasing the expression of target genes such as GLI1 and PTCH1 itself (Figure 5)71.

A similar paradigm is given by two other inherited syndromes, so called Li-Fraumeni and Turcot, containing genetic changes in the TP53 and APC genes, respectively. Li- Fraumeni syndrome is a hereditary disorder with autosomal dominantly inherited mutations of the gene coding for the p53 tumor suppressor, which predisposes affected patients to develop all kind of tumors72,73. Mutant p53 can be conductive for tumor formation due to aberrant cell cycle control and failure of cell cycle arrest, senescence or apoptosis induction74,75. Patients with Turcot syndrome tend to develop multiple colon polyps and malignant brain tumors. Turcot syndrome, also known as “mismatch repair cancer syndrome“ (MMRCS), is characterized by germline mutations of APC or biallelic mutations in DNA mismatch repair genes76,77. Investigation of this inherited syndrome leading to Wnt pathway activation contributed to uncover mutations of CTNNB1, APC, and AXIN1/2 in approximately 15% of sporadic medulloblastomas. The wild-type APC allele has been reported to be lost in a subset of medulloblastoma patients with underlying Turcot syndrome, suggesting that APC acts as a tumor suppressor in the cerebellum. Loss-of-function-mutations in the APC gene result in activation of the Wnt signaling pathway, which plays a major role in the regulation of cell proliferation and differentiation during CNS development as well as in tumorigenesis. ß- catenin is a central effector of the Wnt pathway. The cytoplasmatic amount is controlled by AXIN1 and its homologue AXIN2 (Conductin), which function as Wnt antagonists and interact with APC and glycogen synthase kinase-3ß (GSK-3ß) to form a multiprotein complex. This so-called destruction complex promotes NH2-terminal phosphorylation of ß-catenin by GSK-3ß, marking it for degradation by the proteasome system (Figure 5). Besides genetic changes in the APC gene, which are found particularly frequent in colon cancers, oncogenic missense mutations in the N-terminal part of the ß-catenin gene have been shown in several cancers. This mutant form is resistant to ubiquitination and proteasomal degradation78,79, which leads to increased complex formation of nuclear ß- catenin with the T-cell factor (TCF) resulting in elevated expression of Wnt target factors like Cyclin D1 and cMYC80,81.

15

Introduction

Figure 5 SHH and Wnt signaling pathways. Inactive SHH pathway status: Ptch receptors inhibit Smoothened receptors (Smo, red) to prevent the access of Smo to the primary cilium. Smo receptors could be incororated by endosomes (blue). Gli2 and Gli3 are downstream effectors of the SHH pathway, SuFu binds them. Gli2 is also targeted for degradation via the proteasomal pathway which occurs in the cytoplasm and primary cilium. Active SHH pathway status: Ptch stops to repress Smo (green), which moves into the primary cilium. There, Kif7 aids activation of Gli proteins SuFu releases Gli2 and Gli3, and Gli2 translocates to the nucleus where it functions as a transcriptional activator. Gli3 exhibit repressor actions. Inactive Wnt pathway status: β-catenin, a central effector of Wnt signaling, is bound and phosphorylated in a multi- protein complex consisting of Apc, Axin, CK1, and Gsk3ß. This so-called destruction complex processes ß- catenin for degradation through the proteasomal pathway. Inactive Frizzled receptors are shown in red. Active Wnt pathway status: Wnt binds to Frizzled receptors to activate them (green). The complex is disrupted and allows β-catenin to accumulate and translocate to the nucleus. There it acts as a transcriptional activator. SFRP can bind Wnt to prevent the ligand binding to the receptor. Adapted from Ellison et al.53

1.2.3 Molecular Subgroups of Medulloblastoma Current clinical classification schemes for medulloblastoma include histological sub- grouping according to the WHO classification and clinical markers such as patient age at diagnosis, level of surgical resection, and mestatsic stage at diagnosis. Beyond these clinical characteristics, various research groups have begun to sub-classify medulloblastoma on the basis of genomewide transcriptional and genetic profiling studies to get a deeper insight into the underlying biological pathomechanisms82. This approach revealed distinct molecular subtypes differing in somatic genetic aberrations, transcriptional profiles, demographics, clinical outcome, and histology. A consensus conference in Boston, Massachusetts has taken place in fall 2010 to find an agreement about variations in composition and number of subgroups between different research

16

Introduction

studies. The reached consensus nomenclature defines that four main molecular sub- groups of medulloblastoma exist, which are called Wnt, SHH, Group 3, and Group 4 (Figure 6)83,84. Beside the two subgroups Wnt and SHH, which are named according to the signaling pathways thought to play a driving role in their pathobiology, the other two remaining subtypes are termed Group 3 and Group 4 and often referred to as non- Wnt/non-SHH tumors. These are not obviously characterized by aberrant activation of specific signaling pathways, although there is an association with up-regulation of certain gene classes, such as photoreceptor genes and neuronal genes85,86. The four molecular subgroups are clearly distinct in terms of demographics, histology, cytogenetic aberrations, transcriptome, metastatic stage and clinical outcome. In the following section, the distinctive characteristics are described in more detail for each core subgroup. It is considered to adopt this molecular subgrouping system for more precise identification and outcome prediction of medulloblastoma patients in the clinic. Furthermore it is conductive to identify target cohorts for therapeutic stratification and treatment with certain drugs (e.g. SHH inhibitors).

Figure 6 Overview of the various medulloblastoma subgroups including characteristic attributes. Adapted from Taylor et al.83

17

Introduction

In contrast to tumors belonging to the SHH and Wnt subtypes, which are clearly separated from each other on principal components analysis after unsupervised hierarchical clustering of microarray gene expression data, the non-SHH/Wnt subgroups are not as clearly set aside from each other84, but are still different in a way that separation is justified.

Wnt subgroup Patients harboring WNT-driven medulloblastoma exceed survival rates of about 90%83. Overall, medulloblastoma is more common in boys than in girls, but the gender ratio for WNT tumors is 1:1. WNT medulloblastomas are predominantly found in older children and teenagers (aged 4–15 years), and are rarely present in infants. Up to 25% of adult medulloblastomas can be categorized into the WNT subgroup87,88. Missense mutations in APC and AXIN1 have been identified in sporadic medulloblastoma resulting in overactivity of the canonical Wnt signalling pathway53,89. Additionally, up to 18% of medulloblastomas have been shown to exhibit somatic mutations of CTNNB1 encoding ß-catenin, which accounts for its nuclear accumulation leading to aberrant activation of the Wnt signaling pathway as first described by Ellison and colleagues. This genetic event is now commonly accepted as a hallmark for standard- (or low-) risk medulloblastoma90,91. The etiological role of canonical Wnt signaling in formation of medulloblastoma defines the nomenclature of these tumors as the “Wnt subgroup”. Nearly all tumors belonging to this subtype represent classic histology, regularly show nuclear ß-catenin accumulation and very often come along with monosomy 6 (deletion of one copy of ). Indeed, there are also occasionally tumors showing a transcriptional Wnt signature without having an aberration in copy-number status of chromosome 684. Monosomy 6 is found in most cases of pediatric WNT medulloblastoma, whereas in adult WNT tumors monosomy 6 is evident in only half of cases, and has been also found in adult cases of SHH and Group 4 medulloblastomas. As such, monosomy 6 is a specific marker only for pediatric - and not for adult - WNT medulloblastoma. Metastatic dissemination at presentation is rare in cases of WNT medulloblastoma, and preliminary data suggest that metastatic dissemination may not have a negative prognostic value in this setting87. Despite the known hyperactivity of Wnt signalling in a number of common adult malignancies, e.g. in colon cancer, none of the current therapies that target this pathway have shown to be beneficial in patients with medulloblastoma. Although mutations in the tumor protein 53 (TP53) gene are commonly found in WNT medulloblastoma, agents that target mutant TP53 are not regularly used

18

Introduction

for medulloblastoma therapy. Up to now, no other therapy targets for WNT medulloblastomas are known87. Gibson et al. recently published a medulloblastoma mouse model of the Wnt subtype suggesting that these tumors arise from the lower rhombic lip (LRL) from cells of the embryonic dorsal brainstem92. This model serves as preclinical tool for the assessment of novel therapeutics, as due to the good prognosis of Wnt medulloblastoma, many patients harboring this subtype presumably are overtreated by current therapy schemes. Active discussions about clinical trials of de-escalating therapies for WNT activated medulloblatoma patients are ongoing among experts83.

Sonic hedgehog subgroup Similar to the WNT subgroup, the gender ratio in SHH medulloblastomas is 1:1. SHH medulloblastomas comprise the majority of tumors observed in infants and adults, but are less common in the intervening age groups87. As described in the preceding chapter, some individuals with Gorlin syndrome have a high risk of developing medulloblastoma. This hereditary syndrome is characterized by germline mutations in the PTCH1 tumor suppressor gene. Interestingly, the SHH medulloblastoma subgroup is characterized by deletion of chromosome 9q whereon the PTCH1 gene is located at 9q2284. Goodrich et al. developed a mouse model based on Ptch1 mutation resulting in murine medullo- blastoma93. Other reports describe medulloblastomas being associated with somatic mutations in genes encoding for SHH pathway members, including SMO, PTCH2, SUFU, and GLI269,87. More than 30% of sporadic medulloblastoma show an over- expression of GLI1, demonstrating that the SHH pathway has a major impact in tumorigenesis94. Furthermore, Rausch et al. recently reported on a striking connection between germline TP53 mutation (Li-Fraumeni syndrome) and SHH-medulloblastoma. Based on whole-genome sequencing they uncovered massive, complex chromosome rearrangements. This study revealed a striking link between TP53 mutation and chromotripsis in SHH medulloblastoma. On top of that, they speculated that this phenomenon is a key driver in the tumorigenesis of tumors63. However, the assignment of a tumor to the subgroup by histology is not an easy task. Histologically, the desmo- plastic nodular subtype is restricted to the SHH medulloblastomas, although less than 50% of SHH tumors exhibit this histopathology. Also MBEN is likely to be found in SHH medulloblastomas and the remaining proportion show classic histology. LCA histology has also been reported in the SHH subgroup. SHH is the only subgroup that includes tumors of all four major histological variants87. Metastases at the time of presentation are uncommon. Whereas metastases at presentation are a negative prognostic factor for

19

Introduction

adults with SHH medulloblastoma, there is no evidence that metastases may have a negative prognostic value in infants with these tumors95. The most attractive therapeutic target in this medulloblastoma subgroup is given by the SHH pathway itself. There are several small molecule inhibitors of the SHH pathway, e.g. inhibitors of co-receptor smoothened (SMO), available for clinical trials. Administration of one such inhibitor showed beneficial effects in the beginning96. Unfortunately drug induced resistance to SMO inhibitor treatment occured in both mice and humans. Targeting of additional driver events in combination with SHH inhibition seems to be crucial to avoid drug resistance, as malignant clones that develop resistance to the anti SHH therapy are unlikely to be simultaneously also resistant to other targets. SMO inhibitors are on the way to phase II trials, so SHH medulloblastoma is likely to be the first subgroup to benefit from targeted therapy. Despite reliable and widely accepted as a distinct subgroup, markers for the proper identification of SHH subgroup tumors are still needed87.

For the reason that less is known about the biology of the remaining two medullo- blastoma subgroups, generic names are maintained until a better understanding of the underlying driving mechanisms is achieved.

Group 3 Compared to the other subgroups, Group 3 medulloblastomas have a very poor prognosis. Group 3 tumors are more common in males than in females, and are observed in infancy and childhood, but very rarly in adults. The reason for restriction of Group 3 medulloblastomas to the pediatric age group is unknown. It was suggested that the cell of origin for Group 3 tumors is lacking in adults87. Group 3 tumors are more similar to Group 4 tumors than to SHH or WNT tumors. Regarding chromosomal aberrations, Group 3 medulloblastomas exhibit more frequent loss of both 5q and 10q and gain of chromosome 1q than do Group 4 tumors84. Tumors of this subtype indicate mostly classic histology, although they enclose the majority of LCA medulloblastoma84,85. Additionally, immunopositivity for NPR3 as a Group 3 marker has been suggested as a simple diagnostic tool84. No germline mutations predisposing individuals to Group 3 medulloblastoma have been identified yet87. Group 3 tumors overexpress genes that were identified to normally play a role in photoreceptor cells in the retina, and it is currently unclear how they affect the pathogenesis of this subset. It has also been reported that MYC amplification (but not MYCN amplification) appears to be almost

20

Introduction

exclusively limited to Group 384,85. Hatten et al. described predominant MYC signaling in Group 3 tumors, and therefore suggested to call them the “MYC group” 61. This Subgroup is highly associated with unfavorable prognosis due to frequent dissemination at diagnosis, and a high risk of recurrence. Indeed, the former usage of metastatic stage as a risk factor for poor prognosis identified patient cohorts enriched for Group 3 medulloblastoma84,87. Therapeutic approaches targeting a transcription factor are difficult and small molecule inhibitors of MYC have not been successful in the clinic yet. Up to now, no studies on other targets in Group 3 medulloblastomas have been described. Existing treatment protocols for high-risk medulloblastoma suggest that doses approaching maximal tolerated levels are associated with significant morbidity in these patients87. Pei et al. developed recently a MYC driven mouse model for medulloblastoma that can be used to test therapies for this devastating disease97,98.

Group 4 Despite being the most common medulloblastoma subgroup, the underlying biology of Group 4 tumors is less well understood. No familial syndromes predisposing individuals to Group 4 medulloblastoma have been identified to date. This medulloblastoma sub- group is found across all age groups, although rarely in infants. It accounts for 25% of adult medulloblastomas, often related with a worse prognosis than in childhood cases99. Isochromosome 17q is the most common cytogenetic change observed in Group 4 tumors (66%), although it also appears in Group 3 tumors (26%). Similarly, deletion of chromosome 17p occurs often in both Group 3 and Group 4, but practically never in Wnt subgroup medulloblastomas. Another prominent cytogenetic change characteristic for Group 4 is the loss of the X chromosome, which is seen in about 80% of females having Group 4 medulloblastoma (with a male:female ratio of 2:1 in this subgroup)84,100. Roughly 30% of all medulloblastoma patients have a Group 4 tumor with an intermediate prognosis, comparable to patients with SHH tumors. Histologically, Group 4 tumors usually belong to the classic variant, although cases of LCA histology have been also observed. Only a minority of Group 4 cases exhibits metastases87. Whereas the MYC proto-oncogene protein is highly expressed in Group 3 and WNT medulloblastomas, and MYCN protein is highly expressed in SHH tumors, Group 4 tumors comprise relatively low expression of both MYC and MYCN, apart from a few exceptional cases with MYCN amplification61,84,85. Multiple studies identified a representative gene signature of processes like neuronal differentiation and development in this subgroup, albeit the clinical relevance of this finding is still undetermined84-86. Amplifications of the

21

Introduction

transcription factor gene OTX2 are restricted to Group 3 and Group 4 medulloblastomas, but therapeutics targeting the OTX2 protein have not been described yet87. KCNA1 was suggested as a Group 4 marker for immunohistochemical subgroup affiliation83.

1.2.4 Treatment of Medulloblastoma Up to now, treatment decisions on medulloblastoma patients are still based on clinical parameters such as the histology of the tumor in combination with patient age, presence of metastatic disease at primary diagnosis, and extent of resection. According to these factors, individuals with medulloblastoma are stratified into risk groups predicting distinct response to therapy and prognosis (standard-/low-risk and high-risk). Current multimodal treatment protocols for standard- and high-risk medulloblastoma patients include surgical resection, different intensities of craniospinal gamma irradiation (17-36 Gy to the neuroaxis, 50-55 Gy to the posterior fossa), and varying regimens of chemotherapy101,102. To reduce peri-tumoral edema, glucocorticoids (GC) such as dexamethasone are frequently used peri-operatively in the management of medulloblastoma. Dosages of 1 mg/kg are given once daily for a period of 2 to 3 days before surgical intervention. This course usually produces a remarkable improvement in the symptoms103. Despite therapeutic achievements during the last decades for medulloblastoma patients enabling long-term survival rates of 80% in children stratified to standard-risk disease104 and up to 70% of patients classified to high-risk medulloblastoma105, cure from the malignancy is often accompanied with severe side effects. Many survivors experience marked intellectual and long-term neurological disabilities secondary to both disease and therapy106-109. The increased understanding of medulloblastoma biology and develop- ment of novel therapies in recent years has not effected the systematic use of empirically based treatment regimens, yet. Regarding therapy options, it is necessary to consider that medulloblastoma comprises at least four distinct molecular subgroups with entirely different clinical and biological features84,85,110. Consequently, to minimize neurological damages of the developing brain, especially in infant patients, current treatment approaches need to be complemented or even partly substituted by more specific agents particularly targeting molecular mechanisms of this malignancy as reviewed by Ellison in 201053. Identification of such pathomechanisms and further investigation of their biological context in medulloblastoma are therefore necessary to improve long term effects of patient treatment.

22

Introduction

1.3 OBJECTIVE

Treatment strategies tailored to the clinicopathological and molecular features of medulloblastoma are crucial to avoid over- or undertreatment of patients. Since clinical and histopathological characteristics do not sufficiently reflect the heterogeneous nature of medulloblastoma and accordingly, clinically classified patients might aberrate from the actual molecular risk group, ambitious efforts have been put in the identification of molecular subgroups.

1.3.1 Chromosome 6q Status Defines Medulloblastoma Subgroups In order to provide a more appropriate risk stratification system for medulloblastoma patients, transcriptome-based subgrouping, mutation analyses of distinct candidate genes as well as specific chromosome aberrations have been identified as potential prognostic factors in retrospective cohorts. Pfister et al. recently defined a molecular risk stratification model for outcome prediction based on cytogenetic subgroups111. Array- based karyotyping of 260 medulloblastomas resulted in the following clinical subgroups based on DNA-copy number aberrations: (i) poor prognosis: gain of chromosome 6q or amplification of MYC/MYCN, (ii) intermediate prognosis: gain of 17q or an i(17q) without gain of 6q or amplfication of MYC/MYCN, (iii) excellent prognosis: 6q and 17q balanced or 6q deletion. The overall survival probabilities of medulloblastoma patients regarding chromosome 6q status are shown in Figure 7.

Figure 7 Overall survival probabilities for chromosome 6q status. Adapted from Pfister et al.111

23

Introduction

1.3.2 The Candidate Gene SGK1 The present study aimed at the identification of candidate genes affected by chromosome 6q aberrations in medulloblastoma. Serum-glucocorticoid regulated kinase 1 (SGK1) was identified as a functionally relevant gene in this genomic region. The serum-and glucocorticoid inducible serine/threonine kinase SGK1 belongs to the AGC- kinase family, which is named after its members proteinkinase A, G, and C (PKA, PKG, PKC)112,113. AGC-kinases are known to transduce signals of hormones and mitogens, thereby mediating a wide range of cellular processes like proliferation and differentiation. SGK1 was originally identified in 1993 as a serum- and glucocorticoid-inducible gene in a rat mammary tumor cell line114. The SGK1 transcript encodes a protein of about 50kDa molecular weight, with an evolutionary highly conserved catalytic domain115. The molecular mechanism regulating SGK1 activity has not been fully characterized. Structurally, it shares significant homology with PKA, PKB/AKT, PKC, and p70S6 kinase114. All of these kinases underlie covalent modification by 3- phosphoinositide- dependent kinase (PDK1)116-119, i.e. the conserved phosphorylation site present in the activation loop of these kinases (Thr256 in SGK1) is phosphorylated by PDK1116-119. PDK1 is part of the phosphoinositide 3-kinase (PI3K) signaling cascade, which may be activated by and other growth factors as e.g. hepatocyte growth factor (HGF), insulinlike growth factor-1 (IGF-1), or activation of G-protein coupled receptors, and transduce signals by generating the second messengers PI(3,4,5)P3, and PI(3,4)P2. Activated PDK1 then phosphorylates SGK1 at Thr256, which leads to a change in conformation118. In addition, another phosphorylation site present in a hydrophobic domain located in the C-terminus (S422 in SGK1), the so-called PDK2 site, is also conserved. Mutation of Ser422 to Ala greatly potentiates SGK1 activation leading to constitutive kinase activity at basal levels120,121. Taken together, SGK1 is regulated by the phosphorylation of these two sites, which both are necessary for complete activation (Figure 8).

24

Introduction

Figure 8 PI3K pathway and SGK1 activation. Inactive SGK1 is located in the cytoplasm and could be associated with the plasma membrane. Stimuli such as hormones, and amino acids activate the receptor tyrosine kinases (RTK) or G protein-coupled receptors (GPCR) on the cell surface. PI3Ks are recruited to the membrane in order to phosphorylate phosphatidylinositol (PI) for the generation of phosphatidylinositol(3,4,5)-triphosphates (PIP3). This process is reversible by phosphatase and tensin homologue (PTEN). PIP3s recruit PDK1, which activates SGK1 by binding to the hydrophobic motif (HM) and phosphorylates the activation loop. Modified from122.

Human SGK1 was for the first time described in 1997 as a gene being upregulated in response to changes in cell volume123. It has been shown that the subcellular distribution of SGK1 is dependent on both cell cycle and hormonal regulation, which correlates with the phosphorylation state of the protein124 suggesting that SGK1 expression and activity serves as an integration point for several signaling pathways. SGK1 has significant consensus substrate sites in its catalytic domain that are generally similar to those of the anti-apoptotic kinase PKB/AKT, suggesting some overlap in protein substrates120,121. SGK1 has been shown to phosphorylate glycogen synthase kinase-3 (GSK-3), Raf kinase, and the forkhead family member FOXO3a in vitro 120,125, which are also targets of the AKT protein kinase, although to date the identification of SGK1 substrates in vivo have remained elusive126. AKT proteins include lipid-binding pleckstrin homology (PH) domains at the N-terminus, while the closely related SGK1 lacks this domain127.

SGK1 activity is tightly balanced on three regulation levels: mRNA expression, protein phosphorylation and protein degradation. In contrast to many other kinases, SGK1 transcription is strictly regulated128. Moreover, phosphorylation at two sites is indis- pensable for full kinase activity, and the mRNA transcript, as well as the protein have a very short half-life114,129. Induction of mRNA expression occurs within a couple of minutes

25

Introduction

after an adequate stimulus114, whereas the transcript has a half-life of only about 20 minutes, therefore belonging to the most transient mRNA species114. On transcriptional levels, SGK1 has been shown to be upregulated upon dexamethasone treatment due to activation of the glucocorticoid receptor130. A glucocorticoid response element (GRE) has been identified within the SGK1 promoter114,126,131,132. Since SGK1 transcription is also induced by serum, certain properties of the blood serum are supposed to induce SGK1. Indeed, there is evidence for several factors and hormones influencing SGK1 gene expression, e.g. follicle-stimulating hormone (FSH), transforming growth factor-β (TGF- β), granulocyte macrophage colony-stimulating factor (GM-CSF), fibroblast growth factor (FGF), platelet derived growth factor (PDGF) and tumor necrosis factor-α (TNF-α) were shown to upregulate SGK1 expression133-137. Besides hormones and growth factors, a series of studies revealed that SGK1 is also under control of a variety of environmental stresses, like UV irradiation, heat shock, , and hyperosmotic stress115.

The SGK1 protein has a half-life of less than 30 minutes, which is tightly regulated by polyubiquitination-dependent proteasomal degradation129. Ubiquitinated SGK1 protein is mainly located at the membrane, whereas the non-ubiquitinated protein is found in the cytoplasma, nucleus and the endoplasmatic reticulum (ER)124,138. The sequence encoding the ubiquitination signal is located in the first 60 amino acids in the N- terminus129,139. The E3 ubiquitin ligase Nedd4-2 is involved in this process140. An evolutionarily conserved N-terminal SGK1 variant with enhanced stability and improved function has been described recently, which is characterized by increased half-life due to reduced ubiquitinylation141. The very short half-life of both, SGK1 mRNA and protein leads to the problem, that baseline SGK1 protein is marginally detectable in many conditions despite of high mRNA expression.

Threonine residues in the N-myc downstream-regulated gene 1 protein (NDRG1-Thr- 346/-356/-366) are phosphorylated by SGK1 but not by related kinases142. Therefore these phosphorylation sites of NDRG1 specifically indicate SGK1 activity143.

1.3.3 (Patho-) Physiological Role of SGK1 SGK1 is physiologically expressed in several tissues, including the pancreas, skeletal muscle, liver, heart, placenta, kidney and brain123,144. In the brain, SGK1 mRNA transcripts have been found besides neurons mainly in oligodendrocytes and microglia

26

Introduction

cells145,146. Sgk1 knockout (sgk1-/-) mice are vital and no obvious phenotype due to the gene inactivation has been found147,148. The functional role of SGK1 has been extensively studied mostly with regard to physiological regulation of metabolism, kidney function, , and salt appetite115,149. Furthermore, there are various patho- physiological implications of SGK1 in metabolic diseases (e.g. diabetes mellitus, metabolic syndrome, , diabetic nephropathy150-153, inflammatory diseases (e.g. liver cirrhosis, lung fibrosis, rheumatism), as well as in neurodegenerative diseases (e.g. Chorea Huntington, Morbus Parkinson)154-156. The implication of SGK1 in tumori- genesis has been found in various solid tumors including breast cancer, prostate-, colorectal-, as well as hepatocellular carcinomas157-161. Several reports suggested a functional role of SGK1 in tumor biology by promoting cell survival and cell-cycle progression. A first hint towards affecting cell-cycle progression was the finding that SGK1 translocates from the cytoplasm to the nucleus upon growth factor or hormone stimulation depending on the cell-cycle phase124,162. In breast cancer cell lines, activated SGK1 negatively regulates stress-activated JNK signaling by targeting SEK1163. Moreover, SGK1 contributes to cell survival, cell-cycle progression, and epithelial de- differentiation via MDM2-dependent p53 ubiquitinylation164. In addition, anti-apoptotic effects of SGK1 were shown to be mediated via activation of nuclear factor κB (NF-κB) signaling due to increased degradation of the NF-κB inhibitor IκB165. Furthermore, in kidney cancer cells, apoptosis signaling was shown to be inhibited by interleukin-2 (IL-2) through regulation of SGK1 at transcriptional and post-transcriptional levels166. SGK1 was described to function complementary to the anti-apoptotic kinase AKT in promoting cell survival by directly phosphorylating and inactivating the pro-apoptotic proteins forkhead transcrition factor FKHRL1 (FOXO3a) and BCL2-associated agonist of cell death (BAD)167,168. By phosphorylating FOXO3a in neuronal cells, SGK1 promotes its export from the nucleus and thereby inhibits its pro-apoptotic function125. Moreover, activated SGK1 has been shown to block apoptosis induced by loss of integrin-mediated cell attachment (anoikis169) by preventing de-attachment-induced dephosphorylation of FOXO3a in canine kidney epithelial cells (MDCK)168. SGK1 has been identified as a mTORC1 (mTOR-raptor) substrate. Thus, mTOR may promote G1 progression in part through SGK1 activation and deregulate the cell cycle in cancers through both AKT- and SGK1-mediated p27 phosphorylation and cytoplasmic p27 mislocalization170. A recent report implicated another possible mechanism for the role of SGK1 in cancer formation, in which SGK1 was described as target gene of the WNT-signaling pathway. Thereby, elevated SGK1 phosphorylation was associated with predominant SGK1 localization to

27

Introduction

the cytosol, inactivation of GSK3ß via phosphorylation by SGK1 and finally exclusion of GSK3ß from the nucleus, resulting in increased c-MYC stabilization171. It was demonstrated that SGK1 expression favors the spontaneous development of intestinal tumors in APC deficient mice regarding apc(Min/+)/sgk1(+/+)mice, which exhibit significantly more tumors than apc(Min/+)/sgk1(-/-)mice172. Finally, a study following chemical carcinogenesis, confirmed that sgk1(-/-) mice developed significantly less colonic tumors than their wild type littermates sgk1(+/+)173. Protection from apoptosis and increased chemotherapy resistance upon glucocorticoid treatment has been found in a broad variety of solid cancers including brain tumors. It was shown that dexamethasone treatment inhibits cisplatin and 5-fluorouracil-induced apoptosis and promotes the growth of the malignant cells174. This phenomenon is associated with a glucocorticoid receptor- dependent, transcriptional induction of anti-apoptotic genes, including SGK1, which also protects breast cancer cells from growth factor starvation-induced apoptosis130,175.

1.4 AIMS OF THE STUDY

Integrative DNA copy-number analysis and transcriptome profiling focusing on genes located on chromosome 6q identified SGK1 as a promising candidate gene in medullo- blastoma with clear subgroup-specific expression. This was further substantiated by the immunohistochemical examination of SGK1 in an independent large cohort of medullo- blastoma revealing SGK1 protein expression as a potential new prognostic biomarker. Consequently, functional analyses in vitro were conducted to investigate the implication of SGK1 in medulloblastoma biology and pathogenesis.

28

Materials & Methods

2 MATERIALS & METHODS

2.1 MATERIALS

2.1.1 Antibodies

Target Source Dilution Supplier NDRG1 Goat 1:2500 Abcam, Cambridge, UK

NDRG1 (pT346) Rabbit 1:1000 Cell signaling, Danvers, USA SGK1 Rabbit 1:1000 Abcam, Cambridge, UK SGK1 Rabbit 1:100 Pineda, Berlin, Germany ß-ACTIN Mouse 1:1000 Santa Cruz Biotechnology, Santa Cruz, USA Goat (HRP-coupled) Donkey 1:5000 Santa Cruz Biotechnology, Santa Cruz, USA Mouse (HRP-coupled) Goat 1:5000 Santa Cruz Biotechnology, Santa Cruz, USA Rabbit (HRP-coupled) Goat 1:3000 Cell Signaling, Danvers, USA

2.1.2 Biochemicals and Reagents

Substance Supplier 7AAD, 7-aminoactinomycin BD Bioscience, Heidelberg, Germany ABsolute™ QPCR SYBR® Green Mixes ABgene, Epsom, UK Acrylamide/bis-acrylamide Roth, Karlsruhe, Germany Agarose Sigma Aldrich, Munich, Germany Ammonium acetat 7.5M Sigma-Aldrich, Munich, Germany Ammoniumpersulfate (APS) Roth, Karlsruhe, Germany Ampicillin Roche Diagnostics, Mannheim, Germany AnnexinV binding buffer (10x) BD Bioscience, Heidelberg, Germany AnnexinV-PE BD Bioscience, Heidelberg, Germany Bacto Agar Difco Laboratories, Detroit, USA Bacto tryptone Difco Laboratories, Detroit, USA Bacto yeast extract Difco Laboratories, Detroit, USA Bicinchonic acid (BCA) Sigma Aldrich, Munich, Germany Boric acid Roth, Karlsruhe, Germany Bovine serum albumine (BSA) Sigma-Aldrich, Munich, Germany Bromophenole blue AppliChem, Darmstadt, Germany Chloroform Merck, Darmstadt, Germany

29

Materials & Methods

Complete Mini protease inhibitors Roche Diagnostics, Mannheim, Germany Copper-(III)-sulfate Sigma Aldrich, Munich, Germany Copper-II sulfate Roth, Karlsruhe, Germany Cyanine 3-CTP (Cy3) Perkin Elmer, Boston, USA Cyanine 5-CTP (Cy5) Perkin Elmer, Boston, USA Desoxynukleotid (dNTP)-Set 100 mM Fermentas, St. Leon-Rot, Germany Dimethylsulfoxide (DMSO) Sigma Aldrich, Munich, Germany Dithiothreitol (DTT) Sigma Aldrich, Munich, Germany DNA Loading Dye Fermentas, St. Leon-Rot, Germany DNA Pol Puffer 1 Roche Diagnostics, Mannheim, Germany ECL (plus) substrate GE Healthcare, München, Germany EDTA 25 mM Invitrogen, Karlsruhe, Germany Ethanol Merck, Darmstadt, Germany Ethidium bromide Sigma Aldrich, Munich, Germany Ethylenediaminetetraacetic acid (EDTA) Merck, Darmstadt, Germany First Strand Buffer Invitrogen, Karlsruhe, Germany Formalin/PBS (10% v/v) Sigma Aldrich, Munich, Germany Formamide Merck, Darmstadt, Germany Gene Ruler™ 1 kb DNA ladde Fermentas, St. Leon-Rot, Germany Glycerol Roth, Karlsruhe, Germany Glycine Roth, Karlsruhe, Germany Hydrochloric acid (HCL) Merck, Darmstadt, Germany LPA (linear polyacrylamide) Ambion, Austin, USA Methanol Merck, Darmstadt, Germany NEB buffer 4 (10x) New England Biolabs, Frankfurt, Germany Nuclease free water Ambion, Austin, USA Oligo-(dT)20 primer Invitrogen, Karlsruhe, Germany PE annexin V BD, Heidelberg, Germany Phenylmethylsulfonylfluorid (PMSF) Sigma-Aldrich, Munich, Germany Potassium chloride (KCl) Sigma-Aldrich, Munich, Germany Potassium dihydrogen phosphate (KH2PO4) Carl Roth, Karlsruhe, Germany Propidium iodide Sigma Aldrich, Munich, Germany RNAse-free wate Ambion, Austin, USA SOC medium Invitrogen, Karlsruhe, Germany Sodium acetate (NaAc) Roth, Karlsruhe, Germany Sodium borate Merck, Darmstadt, Germany Sodium chloride (NaCl) Merck, Darmstadt, Germany Sodium citrate Merck, Darmstadt, Germany Sodium dodecyl sulfate (SDS) Sigma Aldrich, Munich, Germany

30

Materials & Methods

Sodium fluoride (NaF) Roth, Karlsruhe, Germany Sodium hydrogenphosphate (Na2HPO4) Sigma-Aldrich, Munich, Germany Sodium hydroxide (10 M) Sigma-Aldrich, Munich, Germany Sodium orthovanadate Roth, Karlsruhe, Germany Spectra multicolor broad range protein Fermentas, St. Leon-Rot, Germany ladder Stratagene human reference RNA Stratagene, La Jolla, USA T4 gene 32 protein USB, Cleveland, USA Tetramethylethylenediamine (TEMED) Roth, Karlsruhe, Germany Tris-base Sigma Aldrich, Munich, Germany Tris-hydrochloride Roth, Karlsruhe, Germany Triton X-100 Sigma Aldrich, Munich, Germany Trizol reagent Invitrogen, Karlsruhe, Germany Tween 20 Sigma Aldrich, Munich, Germany Whole milk powder Roth, Karlsruhe, Germany β-Mercaptoethanol Biorad, Hercules, USA

2.1.3 Buffers and Solutions

Solution Composition Blocking solution 10% milk/3% BSA in TBS-T FACS buffer 0.1%BSA, 0.02%Azide in 1xPBS Laemmli-buffer (5x) 8 % SDS, 200 mM Tris, 0.08 % bromphenol blue, 40 % glycerol, 10 % 2-mercaptoethanol, pH 6.8 LB-medium 10g Bacto tryptone/peptone, 5g Bacto yeast

extract, 10g NaCl, ad 1L dd H2O Nicoletti buffer 0.1% sodium citrate, 0.1% Triton X-100, 50μg/ml PI

Phosphate buffered saline (PBS) 137 mM NaCl, 2.7 mM KCl, 9.2 mM Na2HPO4,

1.8 mM KH2PO4, pH 7.4 Protein-lysis buffer (1x) RIPA buffer, 10 mM NaF, 1 mM sodium orthovanadate, 10 mM phenylmethyl- sulfonylfluorid (PMSF), 1 x Mini Protease Inhibitor Cocktail RIPA buffer (1x) 150 mM NaCl, 1.0% Nonidet P-40, 0.5% sodium deoxycholate, 0.1% SDS, 50 mM Tris, pH 8.0 SDS-PAGE buffer (1x) 25 mM Tris, 200 mM glycine, 0.5% SDS, pH 8.8 TE-buffer (Tris-EDTA) 10 mM Tris-HCl, 1 mM EDTA, pH 8.0 Transfer buffer (1x) 25 mM Tris, 200 mM glycine, 20 % methanol,

31

Materials & Methods

pH 8.8 Tris buffered saline (TBS) (1x) 150 mM NaCl, 10 mM Tris, pH 7.4 Tris buffered saline Tween20 (TBS-T) 150 mM NaCl, 10 mM Tris, 0.05% (v/v) Tween20, pH 7.4 Tris-borate-EDTA (TBE) (1x) 89 mM boric acid, 89 mM Tris-base, 2 mM EDTA,pH 8.0 Tris-Glycine SDS running buffer (1x) 25 mM Tris, 192 mM Glycine, 0.1% SDS, pH 8.3 Western Blot stripping solution 62.5 nM Tris-HCl, 2% SDS, 200 nm 2- mercaptoethanol, pH 6.7

2.1.4 Cell Culture Reagents

Reagent Supplier DMEM (Dulbecco´s Modified Eagle Gibco BRL/Invitrogen, Karlsruhe, Germany Medium); 4500 mg/L glucose Fetal Calf Serum (FCS) Biochrom AG, Berlin, Germany L-glutamine Gibco Invitrogen, Karlsruhe, Germany MEM alpha Gibco BRL/Invitrogen, Karlsruhe, Germany OptiMEM Gibco Invitrogen, Karlsruhe, Germany Penicillin/Streptomycin (1000 U/ml) Gibco Invitrogen, Karlsruhe, Germany Phosphate Buffered Saline Gibco Invitrogen, Karlsruhe, Germany Polybrene Millipore, Billerica, USA Polyethylenimine Polyscience, Niles, USA Puromycin (10 mg/ml) Invitrogen, Karlsruhe, Germany TransIT-293 transfection reagent Mirus Bio, Madison, USA Trypsin/EDTA (0.05%w/v) Invitrogen, Karlsruhe, Germany

2.1.5 Cell Lines and Bacterial Strains

Cell line Origin Acc.-No. Supplier/Source D283 MED Human medulloblastoma HTB-185 ATCC, Manassas, USA Daoy Human medulloblastoma HTB-186 ATCC, Manassas, USA HEK293T Human embryonic kidney CRL-1273 ATCC, Manassas, USA MED8A Human medulloblastoma provided by Michael Taylor, Toronto, USA OneShot TOP10 competent E.coli Invitrogen, Karlsruhe, Germany ONS-76 Human medulloblastoma provided by Michael Taylor,Toronto, USA UW228-2 Human medulloblastoma provided by Steve Clifford, Newcastle, UK UW228-3 Human medulloblastoma provided by Steve Clifford, Newcastle, UK

32

Materials & Methods

2.1.6 Drugs

Substance Supplier 4-hydroxycyclophosphamide (4-HC) Niomech, Bielefeld, Germany Dexamethasone Sigma-Aldrich, München, Germany GSK650394 (by GlaxoSmithKline) Oncology R&D; Collegeville, USA Lomustine (CCNU) Medac, Wedel, Germany

2.1.7 Enzymes

Enzyme Supplier Cla I New England Biolabs, Ipswitch, USA DNase Shrimp recombinant USB Corporation, Cleveland, USA GoTaq DNA polymerase Promega, Mannheim, Germany Not I New England Biolabs, Ipswitch, USA Precisor Polymerase Biocat, Heidelberg, Germany RNase H Qiagen, Hilden, Germany SuperScriptII reverse transcriptase Invitrogen, Karlsruhe, Germany T4 DNA Ligase Roche Diagnostics, Mannheim, Germany Taq DNA Polymerase Qiagen, Hilden, Germany XbaI Fermentas, St. Leon Rot, Germany XhoI New England Biolabs, Frankfurt, Germany

2.1.8 Instruments

Instrument Supplier ABI PRISM 7900 Sequence Detection Applied Biosystems, Foster City, USA System Agilent 2100 Bioanalyzer Agilent Technologies Inc., Santa Clara, USA Agilent DNA Microarray Scanner Agilent Technologies Inc., Santa Clara, USA G25505B Axioskop 40 microscope Carl Zeiss, Jena, Germany Biofuge Fresco Heraeus Instruments, Hanau, Germany EL800 Universal Microplate Reader Bio-Tek instrum., Bad Friedrichshall, Germany Eppendorf Mastercycler Gradient Eppendorf, Hamburg, Germany FACSCanto II BD Biosciences, San Jose, USA Gel electrophoresis power supply E-C Apparatus Corporation, Holbrook, USA Heating block QBT Grant Instruments, Cambridge, UK HMT 702 C microwave oven Robert Bosch GmbH, Stuttgart, Germany Hybridisation Oven Agilent Technologies, Santa Clara, USA Incubator Thermo Forma Thermo Fisher Scientific, Langenselbold,

33

Materials & Methods

Germany Leica DM IRB/E inverted microscope Leica, Wetzlar, Germany system L8-M Ultracentrifuge Beckmann Coulter GmbH, Krefeld, Germany Mini Protean gel electrophoresis BioRad, Munich, Germany system NanoDrop ND-1000 spectrometer NanoDrop, Wilmington, USA Pipettes Gilson, Middleton, USA Sterile bench Hera Safe Thermo Fisher Scientific, Langenselbold, Germany Unimax 1010 Shaker Heidolph Instruments, Schwabach, Germany UV Gel Documentation BioRad, Hercules, USA Vakuumcentrifuge (Concentrator Eppendorf, Hamburg, Germany 5301) Varifuge 3.0 Heraeus Instruments, Hanau, Germany Vi-CELL XR 2.03 Beckman Coulter, Krefeld, Germany Vortex mixer Neo lab 7-2020, Heidelberg, Germany Water bath SW22 Julabo Labortechnik, Seelbach, Germany

2.1.9 Materials

Material Supplier 96well microtiter plates with flat bottom Nunc, Wiesbaden, Germany ABI PRISM optical adhesive covers Applied Biosystems, Foster City, USA Agilent RNA Nano Chip Agilent Technologies, Santa Clara, USA Blotwet gel transfer system Biorad, Hercules, USA Cell culture flasks Corning, New York, USA FACS tubes BD Bioscience, Heidelberg, Germany Falcon transwell inserts (8 μm pores) BD Biosciences, Heidelberg, Germany Falcon tubes (15ml and 50ml) BD Bioscience, Heidelberg, Germany FUJI Medical X-ray films, Super RX FUJIFilm, Tokyo, Japan Gradient gels 4-20% (Tris-glycine); Lonza LONZA, Basel, Switzerland PAGEr* Gold Precast Gels Kryo-tubes (1.8 ml) Nunc, Wiesbaden, Germany Microtiter plates (384 well) Steinbrenner Laborsyst., Wiesenbach, Germany Mini Trans-Blot wet gel transfer apparatus BioRad, Hercules, USA RIPA buffer Sigma-Aldrich, Munich, Germany PCR tubes (0.2ml) Molecular BioProducts, San Diego, USA Pipette tips (10 μl; 20 μl; 100 μl; 1000μl) Starlab, Ahrensburg, Germany Polyvenylidene fluoride membrane Sigma-Aldrich, Munich, Germany (PVDF)

34

Materials & Methods

Reaction tubes Safe-lock (1.5 ml; 2 ml) Eppendorf, Hamburg, Germany RNase ZAP Ambion, Austin, USA Syringe Millex-HA filters 0.45 µm Millipore, Billerica, USA Tissue culture plates (24 well) Nunc, Wiesbaden, Germany Tissue culture plates (6 well) Nunc, Wiesbaden, Germany Whatman paper Sigma-Aldrich, München, Germany

2.1.10 Molecular Biology Kits

Kit Supplier ABsolute QPCR SYBR® Green Mix Abgene, Epsome, UK BigDye Terminator Sequencing kit Applied Biosystems, Foster City, USA CloneJet PCR cloning kit Fermentas, St. Leon-Roth, Germany ECL Western Blot detection kit GE Healthcare, Munich, Germany QiaPrep maxiprep kit Qiagen, Hilden, Germany QiaPrep spin miniprep kit Qiagen, Hilden, Germany QiaQuick gel extraction kit Qiagen, Hilden, Germany Rapid DNA dephos & ligation kit Roche Diagnostics, Mannheim, Germany RNA 6000 Nano kit Agilent Technologies Inc., Santa Clara, USA RNeasy Mini kit Qiagen, Hilden, Germany SuperScriptII RT Kit Invitrogen, Karlsruhe, Germany ViCell XR reagent pack Beckmann Coulter GmbH, Krefeld, Germany Whole Microarray Agilent Technologies Inc., Santa Clara, USA kit, 4x44K

2.1.11 Plasmids and shRNAs

Plasmid/shRNA Supplier MISSION shRNA SGK1 Sigma-Aldrich, München, Germany NM_005627; E7-11; C2; C4 pJet1.2 blunt vector Fermentas, St- Leon-Roth, Germany pLKO.1-Puro TRC 1.5 Sigma-Aldrich, München, Germany pLKO.1-Puro TurboGFP Sigma-Aldrich, München, Germany pLVX-Puro Clontech, Mountain View, USA pLVX-Puro AcGFP Clontech, Mountain View, USA pMD2.G Addgene, Cambridge, USA psPAX2 Addgene, Cambridge, USA

35

Materials & Methods

2.1.12 Primers

Primer Seqence (5´à 3´) ClaI_SGK1_HA AATCGATTCAAGCGTAATCTGGAACATCGTA TGGGTAGAGGAAAGAGTCCGTGGGA DCTN2 fwd CGCCATGGCGGACCCTAAAT DCTN2 rev TTGTCAGCTCCTCCGCATCGAA LMNB1 fwd GCTCCTCAACTATGCTAAGAA LMNB1 rev TCTTTCGAATTCAGTGCTGCTTC NotI_SGK1_fwd GCGGCCGCATGACGGTGAAAACTGAGGC PGK1 fwd AAGTGAAGCTCGGAAAGCTTCTAT PGK1 rev AGGGAAAAGATGCTTCTGGG pJET1.2 seq fwd CGACTCACTATAGGGAGAGCGGC pJET1.2 seq rev AAGAACATCGATTTTCCATGGCAG pLVX-Puro seq fwd CCTGGAGACGCCATCCACGC pLVX-Puro seq rev ACTTGTGTAGCGCCAAGTGC PPIA fwd CGCCATGGCGGACCCTAAAT PPIA rev GCAAACAGCTCAAAGGAGACG SGK1 fwd TGGCACGCCGGAGTATCTCGCA SGK1 rev AAGACAGCTCCCAGGCACCACC SGK1fwd_TQStevens353 GACTGTGGACTGGTGGTG SGK1rev_TQStevens353 CAGGCTCTTCGGTAAACT

2.1.13 Software and Databases

Software/Database Supplier/Source Adobe Photoshop CS2 Adobe, Dublin, Rep. of Ireland Agilent Expert 2100 Agilent Technologies, Santa Clara, USA BD FACS DivaTM BD Biosciences, San Jose, USA BLAST http://www.ensembl.org/Multi/blastview/ http://www.ncbi.nlm.nih.gov/BLAST/ ChipYard© http://www.dkfz.de/genetics/ChipYard/ ClustalW2; http://www.ebi.ac.uk/Tools/msa/clustalw2/ Multiple Sequence Alignment EnsEMBL http://www.ensembl.org/index.html Feature Extraction 9.1 Agilent Technologies, Santa Clara, USA Gap4/ Pregap4 Staden Package MRC Laboratory of Molecular Biology, Cambridge, UK terms http://www.geneontology.org/ GeneCards V3 http://www.genecards.org/ GenePix Pro Version 6.0 Axon Instruments Inc., Union City, USA GraphPad Prism 5.0 GraphPad Software Inc. La Jolla, USA

36

Materials & Methods

KC Junior Bio-Tek, Bad Friedrichshall, Germany KEGG pathways http://www.genome.jp/kegg/pathway.html Microsoft Excel 2007 Microsoft, Redmond, USA Microsoft Powerpoint 2007 Microsoft, Redmond, USA Microsoft Windows Microsoft, Redmond, USA MSigDB v3.0 (Broad Institute’s http://www.broadinstitute.org/gsea/msigdb/ Molecular Signatures Database) index.jsp NCBI http://www.ncbi.nlm.nih.gov/ OligoCalculator http://www.basic.northwestern.edu/biotools/ oligocalc.html PIMS© http://www.dkfz.de/en/genetics/pages/ projects/bioinformatics/PIMS.html PubMed http://www.ncbi.nlm.nih.gov/pubmed PubMed http://www.ncbi.nlm.nih.gov/pubmed/ R 2.2.1 Open source http://www.r-project.org/ R2; microarray analysis and http://hgserver1.amc.nl/cgi-bin/r2/main.cgi visualization platform RankProd http://bioinformatics.oxfordjournals.org/content/2 2/22/2825.full SDS 2.2.2™ Applied Biosystems, Foster City, USA UCSC Genome Browser http://genome.ucsc.edu/

2.2 MOLECULAR BIOLOGY METHODS

2.2.1 DNA Procedures

2.2.1.1 Plasmid-DNA Preparation from E.coli For isolating recombinant generated plasmid-DNA from transformed bacteria (see section 2.2.1.2), plasmid QIAprep kits (spin Miniprep, Maxiprep; Qiagen, Hilden, Germany) were used. Therefore, transformed E.coli cell clones were grown in LB-media containing Ampicillin for selection, over night at 37°C on a bacteria shaker. Following centrifugation at 4000rpm at 4°C, the bacteria pellet was lysed under alkaline conditions according to the manufacturer´s protocol. The alkaline SDS containing buffer from the kit leads to denaturation of bacterial DNA and proteins. After subsequent neutralisation and high salt concentrations, the denaturated genomic DNA and proteins were precipitated and seperated from plasmid DNA by centrifugation. Circular plasmid DNA remained in the supernatant and was purified using colums with silica-membranes. Finally, plasmids were eluated in 50 μl EB buffer and sequenced using Sanger method (see section 2.2.1.3).

37

Materials & Methods

2.2.1.2 Cloning C-terminally HA-tagged SGK1 was generated by amplification of SGK1 (Entrez Gene ID: 6446) from a plasmid pDONR223 kindly provided by Core Facility, DKFZ, Heidelberg. The gene was amplified with Primers NotI_SGK1_for and ClaI_SGK1_HA for including a HA-tag directly in front of the stop-codon using Precisor proofreading polymerase (Biocat) according to manufacturer´s protocol. The calculated primer melting temperature of the region complementary to the SGK1 sequence (irrespective of additional restriction cleavage site and HA-tag) annealing temperature of the PCR was adjusted to 58°C. After amplification in 35 cycles, PCR products were separated by agarose gel electrophoresis and extracted with QIAquick gel extraction kit (Qiagen). Amplicons were inserted into pJet1.2/blunt plasmid using the CloneJet PCR cloning kit (Fermentas). After transformation of OneShot TOP10 competent E. coli cells (Invitrogen), colony-PCR, culturing and plasmid preparation (Qiagen Plasmid Midi Kit), sequences were verified by Sanger sequencing using plasmid-internal primers (pJet1.2 fwd, rev). Subsequently, pJet plasmids containing the SGK1-HA construct and pLVX-Puro plasmid (Clontech, Mountain View, USA) (Figure 9 B) were double-digested with XhoI and XbaI restriction enzymes (10 U each, Biolabs) for 2h at 37°C, enzyme inactivation 15 mins at 65°C. Digested constructs were separated by preparative agarose gel electrophoresis (0.8%) and inserts and linearized target vector were gel extracted and dissolved in water for subsequent ligation. For ligation with T4 ligase (Roche) reaction mix was prepared and vector and insert was added in a ratio 1:3 molecules. Ligation was incubated at 4°C over night and then used for transformation of chemically competent OneShot TOP10 Escherichia coli (E.coli). Subsequent to the cloning procedures, transformed E.coli were grown on selective agar plates containing 100 μg/ml ampicillin. After over night incubation, grown colonies were used for inoculation of Luria-Bertani (LB) medium mini cultures containing 100 μg/ml ampicillin and incubated over night. Plasmids were extracted from cultures using QIAprep Spin Miniprep Kit (Qiagen). The sequence of the cloned gene was verified by Sanger sequencing using plasmid specific primers (pLVX- Puro fwd, rev) and BigDye Terminator v3.1 Cycle Sequencing Kit.

The pLVX-Puro vector is compatible with the lentiviral system of 2nd generation for virus production (see section 2.2.3.2). The transgene SGK1-HA was cloned downstream of the constitutive active CMV promoter located on the pLVX-Puro backbone (Figure 9 B) allowing strong gene expression. The plasmid contains besides the pUC Ori for bacterial

38

Materials & Methods

replication and the resistance gene Ampr for ampicillin selection additionally a viral cis- element between the 5´ and 3´ UTR. RRE (rev response element) binding site enhances virus titers by exporting the RNA of the gene expression cassette outside from the nucleus during virus production. In contrast, the central polypurinetract (cPPT) enhances the import oft he transgene into the nucleus oft he transduced host cell. Moreover, WPRE (woodchuck hepatitis virus posttranscriptional regulatory element) is another regulatory element of the vector supporting processing and maturation of viral transcrips and enhances the export from the nucleus which increases further the lentivirus titers.

Figure 9 Vector maps of plasmids for producing recombinant lentiviruses. A) pLKO.1-puro TRC1.5 plasmid (Sigma-Aldrich) for RNA interfering experiments. B) pLVX-puro vector (Clontech) for transgenic overexpression of SGK1. Adapted from the manufacturer´s protocols176,177.

2.2.1.3 Sanger Sequencing Purified PCR products or plasmid DNA were sequenced using the BigDye Terminator v3.1 Cycle Sequencing Kit (Applied Biosystems, Foster City, USA) including thermally stable AmpliTaq DNA polymerase, modified deoxynucleoside triphosphates (dNTP) and a set of dye terminators labelled with high-sensitivity dyes. 10-20ng purified PCR product or 250ng plasmid DNA at a volume of 6.7μl was employed in a mixture (prepared on ice) containing 3μl BigDye, 0.3μl sequence-specific primer as used for the amplification-PCR

(10μM), and filled up with ddH2O to a final volume of 10μl. After running the sequencing PCR (3 mins initial denaturation at 96°C, followed by 25 cycles of 30 sec denaturation at 96°C, 15 sec annealing at 51°C, 4 mins extension at 60°C), samples were precipitated. The sequencingmixture was added to 1μl 3M sodium acetate (pH 5.2; Roth, Karlsruhe,

39

Materials & Methods

Germany) and 24μl absolute ethanol, centrifuged at 13000rpm for 30min, washed with 200μl ethanol (70%) and centrifuged again at 13000rpm for 15min. After discarding the supernatant, the pellets were dried, resuspended in 10μl formamide (Applied Biosystems, Foster City, USA) and finally sequenced on an ABI PRISM 3100 Genetic Analyzer (Applied Biosystems, Foster City, USA).

2.2.1.4 Array-based Comparative Genomic Hybridization The procedure of array-based comparative genomic hybridization (array-CGH)178 shall be mentioned only in outlines here. Selection of genomic clones, isolation of BAC (bacterial artificial chromosome) DNA, conducting of degenerative oligonucleotide- priming PCR and spotting of PCR products on microarrays were conducted as previously described179,180. Genomic DNA was isolated from tumor tissue as well as from blood of healthy donors using the Qiagen DNA Blood&Tissue Kit (Qiagen). DNA of the tumor sample as well as gender-matched reference DNA (pooled DNA from several healthy individuals) were labeled with different fluorescent color dyes by use of the Bioprime Labeling Kit (Invitrogen). Labeled tumor and reference DNA were precipitated together with unlabeled Cot-1 DNA (Roche) enriched for repetitive DNA sequences in order to suppress cross hybridization events and resuspended in hybridization buffer (ULTRAhybTM, Ambion). After denaturation and re-annealing, hybridization on the array was performed in a GeneTAC Hybridization Station (Genomic Solutions. For post- hybridization washing procedure Tween-20 (Merck) and SSC buffer as well as formamide (Applied Biosystems) was used. Finally, slides were dried by centrifugation (Heraeus) and scanned with an Agilent DNA Microarray Scanner Model G2505B (Agilent Technologies). For image analysis the GenePix Pro 6.1 software (Axon Instruments) was used. Data processing was carried out as previously described179,180.

2.2.1.5 Methylation Array Processing Genome-wide DNA methylation was assessed using the Infinium HumanMethylation450 BeadChip Kit (Illumina, San Diego, USA)181. Methylation analysis of medulloblastoma samples was performed according to the manufacturer’s instructions at the DKFZ Genomics and Proteomics Core Facility (Heidelberg, Germany).

40

Materials & Methods

2.2.2 RNA Procedures

2.2.2.1 RNA Isolation For RNA extraction in order to perform microarray-based expression profiling, 1x107 cells were lysed in 1ml TRIZOL reagent (Invitrogen). After 5 min incubation at room temperature (RT) 300 μl chloroform was added, the sample was mixed thoroughly and incubated for 3 min at room temperature. Subsequently, a centrifugation step for 15 min at 4°C, 13.000 rpm followed. Thereafter, the aqueous phase was collected and mixed 1:2 with 70% ethanol. The solution was applied to Micro RNeasy tubes (Qiagen) for further cleanup. After RNA binding to the column, a washing step with 350μl buffer RW1 (supplied by the manufacturer) followed. Subsequently, a DNase digestion was conducted for 15 min at room temperature with 27U DNase per sample. Thereafter, the RNA was washed again with 350 μl RW1 buffer, 500 μ l RPE buffer, and 500 μl 80% ethanol. Finally, the RNA was eluted in 15-30 μl RNase free water (Ambion) and stored at -80°C until further usage.

For any other experiments, cells were lysed in 350 μl RLT buffer (Qiagen) and homogenized thoroughly. Subsequently, cell lysates were applied to RNeasy spin columns (Qiagen) and total RNA isolation was performed according to the manufacturer’s protocol (RNeasy Mini Kit, Qiagen). Elution from RNeasy spin columns was done with 2 x 30 μl RNase-free water (Ambion) following RNA precipitation for 1 h at -80 °C after adding 1 μ LPA (1 μg/μl), 30 μl ammonium acetate (7.5 M), and 225 μl 70 % ethanol. Finally, centrifugation for 30 minutes at 13.000 rpm, 4 °C and a washing step with 70 % ethanol followed. RNA samples were resolved in 20 μl RNase-free water.

2.2.2.2 Quantification of Nucleic Acids Determination of DNA and RNA concentration in samples was performed via spectrophotometric measurement using a NanoDropTM ND-1000 spectrometer (Wilmington). Absorbance of samples was recorded at wavelength from 220 nm to 350 nm and normalized to values of the corresponding pure solvent. Measurement at 260 nm (A260) was considered to detect DNA/RNA specific absorbance. In accordance with different coefficients for nucleic acid derivatives, an A260 value of 0.1 was equal to 50 ng/μl double-stranded DNA, 40 ng/μl RNA or 33 ng/μl single stranded DNA, respectively. Besides quantification, the absorption spectra characteristics were evaluated for quality

41

Materials & Methods

determination of sample solutions regarding contaminations. Due to aromatic amino acid residues, proteins display a maximum absorbance at 280 nm (A280). Other commonly used substances like EDTA, phenol, and guanidine HCL show considerable absorbance at 230 nm (A230). Calculated ratios therefore indicate a clean solution of DNA and RNA for A260/A280 values of 1.8 and 2.0, respectively, and A260/A230 values between 2.0 and 2.2.

2.2.2.3 Assessment of RNA Quality Total RNA is rapidly degradated for biological as well as chemical reasons. In particular enzymatic digestion of mRNA by various types of RNases alters the transcript composition of samples nonstochastically. Thus, it is of particualr importance to assesss RNA integrity prior to sample preparation for whole transcriptome analyses and qRT- PCR. Therefore, capillary gel electrophoresis on Agilent 2100 bioanalyzer was performed. In principle the RNA is stained with a dye and separated by size. According to the size distribution the RNA integrity number (RIN) is calculated based on values for 18s and 28s rRNA fragments. For this procedure 300-500ng RNA were denatured for 2 min at 70°C. The nano dye intercalating in RNA was mixed with the gelmatrix (both supplied by the manufacturer) and dispensed in the Nano Chip according to the manufacturer´s protocol. Subsequently, 1μl of denatured sample RNA as well as 1μl denatured ladder RNA were applied following RNA analysis by running the “Eukaryotic total RNA” program. Only samples with RINs > 8 were used for further analyses.

2.2.2.4 Reverse Transcription For preparation of cDNA, 1 μg of total RNA from both tumor or cultured cells or Stratagene human reference RNA (Stratagene, La Jolla, USA) was used as a template for reverse transcription with SuperScript II Reverse Transcriptase Kit (Invitrogen, Karlsruhe, Germany) according to the manufacturer’s protocol.

2.2.2.5 Design of Primer Pairs Forward and reverse primers for real-time PCR were designed manually following several aspects. The length of forward and reverse primer was between 20 to 30 base pairs and was designed to be similar in length if possible. The annealing temperature was calculated to be between 59 and 61°C. All primers were designed in an intron

42

Materials & Methods

spanning way to avoid amplification of genomic DNA. The possible generation of primer dimers and stem-loop structures was minimized by running the primer sequences on Oligonucleotide Property Calculator (http://www.basic.northwestern.edu/biotools/oligo- calc.html). To guarantee gene specificity all primers were blasted on NCBI home page (http://blast.ncbi.nlm.nih.gov/Blast.cgi) and in-silico PCR using UCSC Genome Bio- informatics platform (http://genome.ucsc.edu/). The primer pairs were purchased from Sigma-Aldrich.

2.2.2.6 Quantitative Real-Time PCR (qRT-PCR) Quantitative real-time PCR is an established method to determine relative amounts of specific transcripts. It combines a RNA-dependent DNA synthesis, which is performed by a reverse transcriptase enzyme and is therefore termed reverse transcription (RT), with a subsequent PCR (polymerase chain reaction). The PCR on the initially generated cDNA uses specifically designed primers (see section 2.2.2.5) to only amplify the mRNA of a particular target gene and is conducted in the presence of the fluorochrome ABsolute- QPCR SYBR® Green (ABgene). Upon intercalating into DNA double strands, this dye shows a more than 500-fold increase of light emission at 522 nm (λmax=522 nm). The measured fluorescence intensity at λmax therefore directly reflects the amount of amplified DNA and can be examined in real-time to monitor its exponential increase. The amount of transcript-specific DNA products at any reaction cycle is dependent on the initial mRNA abundance of the target gene and the primer efficiencies. The reaction consisted of 6 μl SYBR green, 0.12 μl forward primer (10 μM), 0.12 μl reverse primer (10 μM), 2 μl template cDNA (approximately 100 ng/μl) in a total volume of 12 µl. Each cDNA sample was analyzed in triplicates. Amplification and signal detection was done on an ABI PRISM 7900RT Sequence Detection System (Applied Biosystems). To calculate the efficiency of the PCR a standard curve for every amplicon was measured in every run with stratagene human reference RNA. Initially, the samples were denatured for 15s at 95°C followed by 40 cycles of 15sec at 95°C (denaturation),10sec at 60°C (primer annealing) and 72°C for 60sec. Data were analyzed using the SDS 2.2.2 software (Applied Biosystems). The relative quantification of the RNA of interest in comparison to two housekeeping genes (DCTN2, PGK1) was calculated according to a previously published algorithm182.

43

Materials & Methods

2.2.2.7 Microarray-based RNA Expression Profiling For the expression profiling experiments performed on transduced medulloblastoma cells containing pLKO.1 constructs, double-stranded cDNA was synthesized from extracted RNA via MMLV reverse transcriptase. Amplified fluorescent complimentary RNA (cRNA) was created via T7 RNA polymerase, which simultaneously amplifies target RNA and incorporates cyanine 3- or cyanine 5- labeled CTP with at least a 100-fold RNA amplification rate, and purified afterwards. cDNA synthesis, cRNA synthesis, amplification and labeling were done using the Agilent Quick Amp Labeling Kit. The labeled cRNA samples were then fragmented in fragmentation buffer at 60°C for 30 min before the microarray hybridization. cRNA of cells with SGK1 knockdown and cRNA of control cells transduced with nt shRNA were hybridized to a Whole Human Genome (4 × 44K) Microarray (Agilent Technologies, Wilmington, DE, USA) overnight at 65°C in a Shel Lab hybridization oven (Sheldon Manufacturing, Cornelius, OR, USA). The hybridization slides were washed, stabilized, dried, and immediately scanned using the Agilent DNA Microarray Scanner. The acquired images were processed by the Agilent Feature Extraction Software. All steps were performed according to the Two-Color Microarray-Based Gene Expression (Quick Amp Labeling) Protocol (Publication Number: G4140-90050 v.5.7, Agilent Technologies, Wilmington, DE, USA). Result files containing all relevant raw data from the microarray-based RNA expression profiling experiments were processed using the in-house-developed ChipYard microarray analysis software (http://www.dkfz.de/genetics/ChipYard), and the statistical programming language R.

2.2.3 Lentivirus-based Procedures

2.2.3.1 Lentivirus-Mediated RNA interference (RNAi) Lentiviruses belong to the retrovirus family and are able to stably infect also non proliferating cells due to transferation of a pre-integrationcomlex following reverse transcription oft he RNA genome183. The lentiviral genome consists of regulatory genes (e.g. tat, rev) and accessory genes (vpu, vpr, vif, nef) contributing to efficient reproduction and pathogenesis, whereas three genes are essential for replication: gag (group-specific antigen), pol (polymerase), and env (envelope). Gag encodes proteins fort he viral capsid, pol encodes viral enzymes such as protease, reverse transcriptase, and integrase. Env encodes membrane proteins for the viral envelope to adsorb the host cell. For producing infectious viral particles, essential genes are located on different

44

Materials & Methods

helper plasmids to ensure a safe gene transfer. The main vector contains genes for the integration into the host genome (5´ and 3´ LTR; long terminal repeats) and the packaging signal Ψ (psi). Instead the viral sequences between 5´ and 3´ LTR, an expression cassette coding for the transgene was included184. For silencing of the target gene SGK1, pLKO.1 constructs containing an expression cassette coding for a gene- specific small hairpin RNA (shRNA) were used (Figure 9 A). Using the pLKO.1 lentiviral vector system (2nd generation), self-inactivating, recombinant lentiviruses (SIN viruses) were produced due to a deletion in the 3´ LTR region for inhibiting regulatory activities of both LTRs. The packaging plasmid psPAX2 (Addgene) encodes gag, pol, and rev. The second packaging plasmid pMD2.G (Addgene) encodes the glycoprotein G (vsv-G), thereby pseudotypical, recombinant viruses were produced targeting a wide spectrum of mammalian cells. After stable integration into the host genome, shRNAs were expressed and bind to the complementary endogenous SGK1 transcripts. The resulting dsRNA complexes lead to effective RNAi in the cell (45). This mechanism consists of processed RNA-duplexes by Dicer are integrated in RISC-complexes (RNA induced silencing complex). Thereby, the RNA strands were seperated by RISC and the antisense binds to complementary mRNA of the target gene followed by degradation of the heteroduplexes of mRNA and shRNA by enzymes of the RISC complex.

2.2.3.2 Production of Lentiviral Particles Lentiviral particles were produced by co-transfection of HEK293T cells (passage <10) with the psPAX2, pMD2.G, and pLVX-Puro or pLKO.1 constructs (MISSIONTM TRC-Hs 1.5)185 (Figure 9). 4x106 cells were seeded into 10cm cell culture plates in high glucose DMEM medium (Gibco) supplemented with 30% FCS (v/v), 1% penicillin/streptomycin (v/v) and 1% 200mM L-glutamin (v/v) and cultivated till reaching 70% confluency at 37°C and 5% CO2 in the incubator for 24h. After exchanging medium (5ml), co-transfections were carried out using 2µg of each packaging plasmid and 4µg of pLVX-Puro or pLKO.1 constructs, respectively. Therefore, 600µl OptiMEM medium with 30µl TransIT® Transfection Reagent (Mirus, Madison, USA) (5 mins incubation at room temperature) were mixed with the plasmids and incubated 20 mins at room temperature to allow forming transfection complexes. Subsequently, the mixture was dropped carfully onto the HEK293T cells, another medium exchange occured after 24h following viral production for another 24h (first harvest) and 48h (second harvest). Virus was harvested and concentrated at 48h and 72h after co-transfection. For increasing yields, upto three

45

Materials & Methods

plates were performed for each construct. Before virus concentration, GFP transfection efficiency was checked under the microscope. The transfected cells looked rounder and should be GFP positive. The medium slightly changed color, an indication of active cell metabolism. Using a 1ml pipette, the medium containing the lentiviruses was carefully collected from transfected plates without disturbing the HEK293T monolayer. Using a 0.45 μm syringe filter (SFCA filter, Corning), medium was filtered into ultraclear SW41 centrifuge tubes (Beckman). Filling each tube till 3-5mm from the top with liquid and balancing is important before ultracentrifugation using a SW41 rotor for 90 mins at 4°C at 25.000 rpm. After centrifugation, the supernatant was carefully decanted into the waste bottle and virus pellet slightly dried. 100 μl sterile PBS or OptiMEM medium (Sigma) was added to the pellet and left standing covered at 4°C overnight. Finally, viruses were resuspended and 10µl stored as 10µl aliquots at -80°C.

2.2.3.3 Determination of Virus Titer and Multiplicity of Infection (MOI) The effective titer of produced transduction units and the multiplicity of infection (MOI) was determined by transduction of HEK293T cells or medulloblastoma cells, respec- tively. Therfore, lentiviral particles containing a GFP cDNA construct were carried with every virus production to measure titer and efficiency afterwards. Consecutive flow cytometry analysis was used for detection of GFP-positive cells. Calculation of virus titer: Titer [TU/µl] = (N*(P/100)*DF)*1/V TU: transduction units; N: cell number at transduction time point; P:percentage of GFP- positive cells; DF: dilution factor; V: volume

2.3 CELL BIOLOGY METHODS

2.3.1 Cell Culture Procedures

2.3.1.1 Cell Culture All cell culture procedures were done under sterile conditions using a laminar flow workbench. The human medulloblastoma cell lines Daoy, MED8A, ONS-76, UW228-2, and UW228-3 were grown in Dulbecco's Modified Eagle Medium (DMEM; Invitrogen) supplemented with 10% fetal calf serum (FCS) and 1% penicillin/streptomycin (Gibco). D283 MED cells were grown in MEM alpha (Gibco) supplemented with 10% fetal calf serum (FCS) and 1% penicillin/streptomycin (Gibco). The normal culture conditions were

46

Materials & Methods

an adherent growth mode in poly-L-lysine-coated flasks (75 cm2, Nunc). All cell lines were grown in a humidified incubator at 37°C in 5% CO2/ 95% O2 and were routinely tested for authenticity and contamination186. For subculturing routineously every 3rd to 4th day, cells were washed once with trypsin/EDTA (0.25%, 1 mM, Invitrogen) and then incubated again with trypsin/EDTA until detachment of cells occured. Following re- suspension in DMEM and centrifugation, cells were resuspended in DMEM and seeded at ratios 1:5 to 1:15. For seeding of cells in appropriate densities cell number was determined using a Vi-CELL XR 2.03 cell counter (Beckman Coulter).

Stock solutions of dexamethasone (Sigma-Aldrich) were prepared by dissolving the pure compound in 100% ethanol in order to achieve a primary concentration of 2.5 mM. The SGK1 antagonist GSK650394 was developed and kindly provided by GlaxoSmithKline (Oncology R&D; Collegeville, PA, USA) and stock solutions were prepared by dissolving the compound in DMSO at a concentration of 10 mM. Working dilutions were achieved by diluting the stock solutions directly in the appropriate cell culture medium. Prior to the administration of dexamethasone, all cells were grown for three days in medium containing charcoal-stripped FCS (Biochrom, Berlin, Germany).

2.3.1.2 Cell Viability and Counting The viability and absolute number of cells were determined with a Vi-CELL XR 2.03 cell counter from Beckmann Coulter. Cells were automatically stained with trypan blue to exclude dead cells from the measurement and average number of cells per ml was calculated.

2.3.1.3 Transduction of Medulloblastoma Cells Transduction was performed in either 6 well plates or 24 well plates, seeding 1x105 or 5x104 cells, respectively. Following attachment and reaching 70-80% confluency after 24h, stable transduction with recombinant, lentiviral particles containing a vector system with an expression cassette coding for either a gene-specific shRNA (pLKO.1) or SGK1- HA transgene (pLVX-Puro) (see section 2.2.1.2) was conducted to induce SGK1 knockdown or overexpression, respectively. Infections of medulloblastoma cells (Daoy, UW228-3) with an MOI of 10, were carried out in the presence of 8 μg/ml of polybrene and virus- containing supernatant was removed 20-24h post transduction. For gene silencing, five independent shRNA constructs targeting different regions of the SGK1

47

Materials & Methods

mRNA transcript were used (MISSION shRNA NM_005627; E7-11; Sigma-Aldrich, München, Germany). Non-targeting shRNA was used as a control. For overexpression of SGK1, pLVX-Puro vectors were used, and empty vectors were used as a control. Calculation of required virus volume: Virus volume [μl]=(N*MOI)/titer [TU/µl] N: cell number at transduction time point; MOI: multiplicity of infection; TU: transduction units

To determine transduction efficencies, cells were transduced with GFP-containing viruses as a positive control for infection. Stable gene silencing was verified by qRT-PCR and Western blotting. Subsequently, diverse functional assays and analyses were performed using always freshly transduced medulloblastoma cells.

2.3.2 Functional Analyses

2.3.2.1 Viability and Proliferation Assay To assess the viability and growth rates of medulloblastoma cells in vitro under various treatment conditions, the MTS [3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)- 2-(4-sulfophenyl) 2Htetrazolium] based CellTiter 96 AQueous Non-Radioactive Cell Proliferation Assay (Promega, Madison, WI, USA) was used following the manufacturer´s recommendations (100µl medium + 20µl MTS/PMS, incubation 3h or 4h at 37°C in a 5%

CO2 humidified incubator. MTS supplemented with phenazine methosulfate (PMS) is converted by metabolically active cells into a water-soluble formazan product with the absorbance quantity at 490nm which is detecable by ELISA-reader. The absorbance at 490nm is proportional to the number of living cells in the test volume187. All samples were assayed in three or six technical replicates and experiments were performed at least twice. The half maximal effective concentration(EC50) of GSK650394, 4-HC and CCNU was calculated for each cell line separately using the GraphPad Prism 5.0 software (GraphPad Software, La Jolla, USA).

2.3.2.2 Chemotherapy Experiments CCNU (lomustine) and cyclophosphamide are alkylating agents used in cancer chemotherapy. Clinical applied as a prodrug, cyclophosphamide is metabolized in vivo by cytochrome P450 enzymes mostly in the liver188,189 to generate 4-hydroxycyclo-

48

Materials & Methods

phosphamide and its tautomer aldophosphamid which further dissociates into acrolein and the actually active phosphoramide mustard190-192. Therefore, in vitro experiments were conducted by use of an already activated form of cyclophosphamide, namely 4- hydroperoxycyclophosphamide (4-HC) which is converted into 4-hydroxycyclophospha- mide without enzymatic involvement193,194.

Medulloblastoma cell suspension was diluted to 4x104 cells/ml, 4000 Daoy and UW228-3 cells were seeded in 96 well microtiter plates in a volume of 100 μl and were allowed to attach for 24h. 4-HC (Niomech, Bielefeld, Germany) and CCNU (Medac, Wedel,

Germany) purchased as powder, were dissolved immediate before application in ddH2O and absolute ethanol, respectively and diluted with the respective solvent and medium to the designated concentrations. Chemotherapeutics were applied at final concentrations ranging from 0.5μM to 40μM for 4-HC and from 5μM to 200μM for CCNU and cells were incubated for another 48h at 37°C in a 5% CO2 humidified incubator. At the designated timepoint MTS assay was performed according to section 2.3.2.1. using an incubation time of 4h.

2.3.2.3 Irradiation Experiments Transduced medulloblastoma cells grown in either 96 well-plates for MTS assay or 24 well-plates for apoptosis assay were exposed to ionizing radiation (IR) under using a Caesium-137 source of radiation at a dose rate of 0.512 Gy/min. IR doses ranging from 2-10 Gy were determined to show significant effects on cell viability after 48 to 120h. As negative control (0 Gy), transduced cells were carried to the radiation source but finally not put into the irradiation chamber.

2.3.2.4 Flow Cytometry Fluorescence activated cell sorting (FACS) is a method for separating microscopic particles like cells in a stream of fluid and passing them by a detection apparatus to determine individual features. FACS examines optical characteristics of cells, for example indicating size (forward scatter; FSC) or granularity (side scatter; SSC) detectable as light scattering. In particular, measurement of fluorescence intensities enables this technique to quantify any labeled component of an individual cell and hence to compile various distributions of cellular features within a given population. Flow

49

Materials & Methods

cytometry analyses reported in this study were performed on a FACSCanto II flow cytometer (BD Bioscience) using BD FACS Diva software (BD Bioscience).

2.3.2.5 Cell Cycle Analysis and Detection of SubG1-fractions Cell cycle analyses are generally distinguished from proliferation assays as they provide a snapshot of the cell cycle with quantitative information about cell proportions residing in different phases. Propidium iodide (PI) is a DNA-intercalating substance being quanti- tatively detectable by fluorescence measurements. Accordingly, using FACS analysis, stained cells can be discriminated between a normal diploid karyotype, representing G0 or G1-phase, and a tetraploid karyotype, indicating G2-phase or mitosis. Apoptotic cells are characterized by a hypoploid DNA content and discriminable from cells with normal (diploid) DNA content, thus represented as subG1-fraction. Besides the two distinct distribution peaks of G0/G1 and G2/M, a transient intermediate state can be identified representing S-phase. The relative amount of cells being in G0/G1 or G2/M at the time point of preparation can reflect overall difference in proliferation behavior or distinct imbalances in checkpoint control and thus provides a suitable overview of the cell cycle. For detection of cell cycle phases after medulloblastoma cell treatment, Nicoletti assay was performed195. Transduced medulloblastoma cells were stained with PI by incubation in hypotonic buffer containing 50μg/ml PI (Propidium iodide, Sigma Aldrich, Munich, Germany), 0.1% sodium citrate and 0.1% Triton X-100 (Sigma Aldrich, Munich, Germany) for 4h at 4°C in the dark. Nicoletti buffer was applied 1:1 (v/v) directly into the well where cells are grown. Due to the hypotonic propensity of the buffer, cell nuclei were extracted and subjected to FACS measurement using FACSCanto II (BD Biosciences). For each measurement, at least 20.000 events were counted. Nuclei fluorescence after laser excitation was used to quantify the distribution of cells in each cell-cycle phase: subG1, G1, S and G2/M. Analysis was evaluated using FACSDiva software (BD Biosciences).

2.3.2.6 Apoptosis Assay Phosphatidylserine externalization as a measurement for early apoptosis was analyzed by flow cytometry. To quantify apoptotic cells, double stainings with Annexin V-PE and 7- amino-actinomycin (7-AAD) were conducted. Therfore, 1x105 to 1x106 transduced or drug treated medulloblastoma cells were harvested by centrifugation. Subsequently, cells were incubated with 30μl of an Annexin V-PE/7AAD solution (1:10 Annexin V-PE/7AAD

50

Materials & Methods

in 1x annexin binding buffer) for 15min at 4°C in the dark. Directly after incubation 150μl of 1x annexin binding buffer was added, immediately followed by flow cytometry using FACS Canto II (BD Biosciences). For each measurement, at least 10.000 events were counted. Data were analyzed using the FACS Diva software (BD Biosciences).

2.3.2.7 Migration Assay An well-established method to determine differential numbers of migrated cells under various conditions is provided by the Boyden chamber assay. The assembly comprises an upper compartment where the cells are seeded in cell culture medium, and a lower compartment containing culture medium supplemented with a diffusible migration stimulus. Both compartments are separated through a porous membrane allowing the stimulus to spread and attract cells to actively pass. The pores size approximately constitutes half the cell diameter and therefore favors an active movement towards the increasing attractant gradient over a sporadic motility. The relative amount of cells residing in the lower compartment, after incubation in the presence of an adequate stimulus, gives an indication of different migration properties. In the present study, a migration assay using 24-well plates with transwell inserts (8 μm pores, BD Falcon) was applied to adherent medulloblastoma cells (Daoy, UW228-3) transduced with pLVX- SGK1-HA or pLVX-empty vector constructs, respectively. 50.000 cells were seeded into the inserts (upper compartment) containing medium with 0.5% serum, whereas the lower compartment contained medium with 10% serum as a migration stimulus. Cells were allowed to migrate 24h at 37°C and 5% CO2. Subsequently, transwell inserts were taken off and cells at the membrane, facing the upper compartment, were thoroughly removed with a cotton wool stick. Cells at the bottom of the membrane were washed with PBS and fixated for 15 minutes in 5% PFA. Afterwards, membranes with fixated cells were rinsed with PBS and the membrane was cuttet out from the well. Thereafter, the membrane was mounted on glass slides using ProLong® Gold mounting medium containing DAPI nuclear stain (Invitrogen, Karlsruhe, Germany). Finally, stained nuclei were counted at the microscope.

51

Materials & Methods

2.4 PROTEIN BIOCHEMISTRY METHODS

2.4.1 Protein Procedures

2.4.1.1 Isolation of Protein For extraction of total protein of cultured cells, 1x106 medulloblastoma cells were pelleted by centrifugation and washed with PBS prior to addition of 40-70µl RIPA-lysis buffer. RIPA buffer (Sigma-Aldrich) was supplemented with 10 mM NaF, 1 mM sodium orthovanadate, 10 mM phenylmethylsulfonylfluorid (PMSF), and complete Mini Protease Inhibitor Cocktail Tablets (Roche) for Ser/Thr phosphatase and protease inhibition.

2.4.1.2 Protein Concentration Measurement by BCA Assay Protein was quantified using a colorimetric assay based on reduction of Cu2+ by peptide bonds to Cu+ ions under alkaline conditions, which is then chelated by bicinchoninic acid (BCA) and exhibit a high absorbance at 562 nm. Thus, 40 µl 0.1% copper-II-sulfate in BCA was added to 2 µl of protein solution and incubated for 30 min at 37°C. Absorption was measured and protein concentration was derived from a standard series generated with bovine serum albumine (BSA).

2.4.1.3 Western Blot Analysis Size-dependent, electrophoretic separation of protein fractions were performed with SDS-PAGE (sodium dodecyl sulfate-polyacrylamide gel electrophoresis). Therefore, gels of 1 mm thickness were prepared using the Mini-PROTEAN system (BioRad) by successively casting separation (10 % acrylamide, 375 mM Tris, pH 8.8, 1% SDS, 0.1% APS (ammoniumpersulfate) and 0.01% TEMED (N,N,N’,N’-tetramethylethylenediamine)) and stacking gel (5% acrylamide, 125 mM Tris, pH 6.8, 0.1% SDS, 0.1% APS and 0.01% TEMED). Alternatively, gradient gels (Lonza PAGEr* Gold Precast Gels, LONZA) were used together with Tris-Glycine SDS running buffer (25 mM Tris, 192 mM Glycine, 0.1% SDS, pH 8.3. Protein extracts were adjusted to the maximum equal concentration. For gel loading 5 x Laemmli buffer (8% SDS, 200 mM Tris, pH 6.8, 0.08% bromphenol blue, 40% glycerol, 10% 2-mercaptoethanol) was added to 40µg total protein and denatured for 5 min at 95°C. For size estimation, Spectra multicolor broad range ladder (Fermentas) was loaded on the gel together with protein samples. SDS-polyacrylamide

52

Materials & Methods

gel electrophoresis (SDS-PAGE) was performed in SDS running buffer with an initial step of 5 min at 65 V following 60-80 min at 130 V. Transfer onto a polyvinyliden flouride (PVDF) membrane was performed in a tank blotting procedure using a Mini Trans- Blotwet gel transfer system (Biorad) and a transfer buffer composed of 25 mM Tris, pH 8.8, 200 mM glycine, and 20 % methanol with stepwise increasing current (10 min 100 mA, 10 min 200 mA, 10 min 300 mA, 10 min 400 mA and 20 min 500 mA). The whole blotting procedure was carried out under permanent ice cooling to prevent heating of the buffer due to high currents. After completed transfer membrane was washed briefly in TBS-T (150 mM NaCl, 10 mM Tris, and 0.1% Tween-20, pH 7.4) and blocked in 10% milk/3%BSA in TBS-T for 1 h at room temperature. Protein detection was done using antibodies in the recommended dilutions, given in section X in 1:10 blocking solution at 4°C over night. After washing 3 x 5 min in TBS-T and incubation with horse- radish peroxidase (HRP) coupled secondary antibodies for 1 h at room temperature, the membrane was washed and subjected to chemiluminescent detection using ECL or ECLplus (GE Healthcare). X-ray films (FUJI) were exposed to stained blots in a developing cassette (exposure time individually adjusted). For subsequent detection of different epitopes on the membrane, bound antibodies were removed by incubation at 52°C in stripping buffer (6.25 mM Tris pH 6.5, 2% SDS, 0.8% ß-mercaptoethanol) for 30 min followed by 1 h blocking.

2.4.2 Immunostaining Prodedure

2.4.2.1 Immunohistochemistry (IHC) A custom tissue microarray (TMA) of pediatric medulloblastoma samples was constructed for investigation by immunohistochemistry. Hematoxylin and eosin (HE)- stained sections from all paraffin blocks were prepared to identify representative tumor regions as previously described196. In addition, samples of non-neoplastic cerebellar tissues were prepared as a control. Immunohistochemistry was performed based on an indirect streptavidin-biotin method. 10 μm sections of the recipient block were prepared, deparaffinized and epitopes were retrieved by immersion of tissue sections in pre-heated 10 mM citrate buffer (pH 6.0) and maintained heating in a microwave oven for 10 min. Endogenous peroxidase activity was inhibited by 3% hydrogen peroxide. Primary anti- human antibody against SGK1 (Abcam, Cambridge, UK) was used. Biotinylated anti- rabbit immunoglobulin served as secondary antibody. Sections were incubated with horseradish peroxidase-conjugated streptavidin for 15 min to increase the sensitivity of

53

Materials & Methods

the detection system. For color development, the specimens were incubated with 3,3'- diaminobenzidine hydrochloride (DAB) supplemented with hydrogen peroxide and afterwards counterstained in hematoxylin. Antibody diluent, secondary antibodies and detection system were used according to the manufacturer’s instructions (Dako, Glostrup, Denmark). Scoring of antibody staining cytoplasmic and nuclear immuno- reactivity were scored separately according to staining intensity and graded semi- quantitatively by neuropathologists. For statistical analyses, the immunostaining classifi- cations were reduced to two categories: low and high expression of the evaluated antigens of interest. Estimation of survival time distributions was performed by use of the Kaplan-Meier analysis. For comparisons of survival curves, the log rank test was used and univariable Cox proportional hazards regression models were applied to examine the association of molecular markers with the hazard of death from disease and disease progression. To account for multiple testing, p-value adjustment was calculated.

2.5 STATISTICAL ANALYSES

Kaplan-Meier survival curves were compared by the Cox-Mantel log-rank test in Winstat for Excel (R. Fitch Software, Germany). P-values were calculated with a two-tailed student´s t-test and ANOVA (analysis of variance) was performed using Microsoft Excel or GraphPad Prism 5.0 software, respectively.

54

Results

3 RESULTS

3.1 IDENTIFICATION OF SGK1 AS CANDIDATE GENE IN MEDULLOBLASTOMA

Previous studies based on DNA copy-number aberrations demonstrated that chromo- some 6q status defines subgroups of medulloblastoma patients with distinct clinical outcome111. Array-CGH analysis demonstrated balanced 6q copy-number status in 44 of the 64 investigated medulloblastoma samples (69%). A chromosomal gain of 6q was shown in 5 tumors (8%). 15 medulloblastomas (23%) exhibited loss of chromosome arm 6q. In order to identify potential candidate genes located on chromosome arm 6q, which are affected by respective changes in gene expression, microarray-based transcriptome analysis was performed. To identify genes differentially expressed between the different subgroups, pooled RNA of five medulloblastoma samples carrying 6q gains were co- hybridized with pooled RNA of five medulloblastoma samples showing a monosomy of chromosome 6. The top 50 upregulated genes at chromosome 6q obtained from this approach are listed in Supplementary Table 1 (Appendix).

3.1.1 SGK1 mRNA Expression is correlated with Chromosome 6q Copy-Number Status in Medulloblastoma In an attempt to further investigate the interrelation between gene expression and 6q copy-number status, transcriptome analysis was performed for each of the tumor samples investigated by array-CGH. This analysis revealed several interesting candidates after ranking the most differentially expressed genes according to their differences comparing the mean values of medulloblastoma harboring a 6q gain vs. tumors with 6q loss. The complete gene list of the top 50 candidates is shown in Supplementary Table 2 (Appendix). Interestingly, SGK1, one of the top-ranking genes among the identified candidates to be upregulated in medulloblastomas possessing 6q gain, was also among the top-ranking genes in the initial expression profiling experiment described above. The respective clones on the microarrays represented the transcription start site of serum- and glucocorticoid regulated kinase (SGK1) at chromosome locus 6q23.2. This gene encodes a serine/threonine protein kinase that plays an important role in cellular stress response and is a crucial member of frequently deregulated pathways in medulloblastoma. For medulloblastoma samples for which a sufficient amount of RNA

55

Results

material was available (n=41), the results derived from the expression profiling experiment Figure 10, left panel) were validated by quantitative real-time PCR (QRT- PCR) (Figure 10, right panel). A high correlation of SGK1 mRNA expression levels with copy-number status of chromosome arm 6q was shown. The positive correlation was stronger than an expected gene-dosage effect (mean log2-ratio of 2.22 in tumors with 6q gain vs. mean log2-ratio of -2.79 in tumors exhibiting 6q loss when compared to normal cerebellum; p < 0.0001), suggesting a functional role of this kinase in tumor biology of the different subgroups.

Figure 10 Expression levels of SGK1 mRNA in primary medulloblastoma. SGK1 expression values of 64 medulloblastoma samples derived from microarray transcriptome analysis were grouped according to chromosome arm 6q copy number status (left panel). These results were validated in a subset (n=41) of tumors from which sufficient amounts of RNA were was available, using quantitative real-time PCR (right panel). SGK1 mRNA expression levels are shown as log2-ratios when compared to normal cerebellum (pool of non-neoplastic cerebellar tissue from 24 male/female Caucasians). Statistical significance was assessed using unpaired t-tests. The experimental analyses were performed by Prof. S. Pfister et al. in collaboration with D. Sturm, DKFZ, Heidelberg).

3.1.2 SGK1 is Differentially Expressed Between Medulloblastoma Molecular Subgroups In order to investigate SGK1 mRNA expression levels according to molecular consensus subgroups as suggested by Taylor et al.83, our data set (n=64) was enlarged (n=152) and combined with recently published medulloblastoma transcriptome studies by Kool et al.85 (n=61), Fattet et al.197 (n=55), and McCabe et al. (unpublished) (n=34) to a total number of 302 medulloblastoma samples, accessible via the microarray analysis and visualization platform ‘R2’. Annotation with genetic aberrations and clinical factors demonstrated monosomy 6 being exclusively present in WNT-driven medulloblastomas and 6q gain was restricted to Non-WNT/Non-SHH tumors. Expression levels of SGK1 mRNA were distinct in subgroups, displaying high expression in variants with molecular

56

Results

and clinical high-risk factors (Group 3 and 4), whereas low expression was detected in cases with excellent and intermediate prognosis (WNT and SHH, respectively, Figure 11 A). This subgroup-specific expression patterns revealed that high SGK1 expression levels were largely limited to Group 3 and 4, thus indicating that it constitutes an excellent marker gene for non-WNT/non-SHH medulloblastoma.

3.1.3 SGK1 Protein Expression is correlated with Overall Survival of Medulloblastoma Patients Furthermore, protein levels of SGK1 were evaluated in an independent cohort of 260 medulloblastoma samples represented on a tissue microarray (TMA) using immuno- histochemistry (IHC). SGK1 positivity was detected in 26% of samples, whereas the remaining 74% of the tumors disclosed a negative or weakly positive immunostaining. Survival analysis by Kaplan-Meier analysis revealed a significant association of SGK1 immunopositivity with poor overall survival (p = 0.0168, Figure 11 B), indicating SGK1 as a potential prognostic marker for high-risk medulloblastoma.

Figure 11 A) Subgroup-specific gene expression patterns of SGK1 in medulloblastoma. mRNA expression profiles were accessible via the microarray analysis and visualization platform ’R2’. Untrans- formed, non-normalized SGK1 gene expression values of medulloblastoma samples (n=302), initially generated on Affymetrix gene expression arrays from different data sets were combined. Statistical significance was calculated using one-way ANOVA (p<0.0001). B) Survival analysis of medulloblastoma patients according to SGK1 immunohistochemistry. Overall survival probabilities for negative or strong (mainly nuclear) staining in medulloblastoma (n=217) were estimated from the the time point of diagnosis using Kaplan-Meier analysis. Medulloblastoma patients with high SGK1 protein expressing tumors showed a significant association with poor survival. The p-value of the analysis is given (p=0.0168). This experiment was performed by Prof. S. Pfister et al. in collaboration with Dr. M. Remke; the evaluation by Prof. A. Korshunov, DKFZ, Heidelberg.

57

Results

3.1.4 SGK1 is Differentially Methylated Across Medulloblastoma Subgroups To further investigate alternative mechanisms explaining subgroup-specific SGK1 expression patterns beyond a gene-dosage effect, PCR based analyses were performed in order to detect the abundance of an evolutionarily conserved N-terminal SGK1 mRNA variant with enhanced stability and improved function. This alternate transcript of SGK1 encodes an isoform, which has been described to be more stable, less susceptible to ubiquitinylation and which was observed to possess improved physiological propensity of epithelial conductance compared to other SGK1 isoforms141. However, upregulation of this specific transcript did not seem to play an essential role in elevating SGK1 activity, as the relative expression in our cohort was negligible comparing it to the overall expression of other SGK1 transcripts (data not shown). In tumors with 6q loss, the alternative transcript could be marginally detected and no significant differences were observed between tumors with 6q gain or balanced 6q. A complementary approach was taken by screening for activating mutations in SGK1, which could provide an alternative mechanism of abnormal SGK1 regulation by creating a constitutively active form. For this purpose, all exons of SGK1 were investigated by genomic sequencing of 25 medulloblastoma samples and corresponding germline DNA of these patients. This analysis revealed no mutations in SGK1 comparing medullo- blastoma vs. matched germline samples.

To account for epigenetic regulatory events governing SGK1 mRNA expression, a bisul- fite conversion-based analysis of medulloblastoma samples (n=115) was conducted using the Human Methylation 450K Array from Illumina. This method allowed monitoring of more than 485.000 potential methylation sites per sample at single-nucleotide reso- lution. Also CpG sites outside of CpG islands and coding regions were covered by this approach. While 5’promoter methylation was virtually absent across all medulloblastoma samples, focusing on the methylation status of CpG sites in the SGK1 locus (gene body and 3’UTR) revealed clear subgroup-specific differences. Significantly higher methylation was found in the WNT and SHH-subgroups, whereas almost no gene body methylation was observed in Group 3 and Group 4 tumors, as shown in Figure 12. The result shows a highly significant negative correlation of SGK1 mRNA expression levels and extent of methylation of several CpG sites clustering together in the 3’ end of the SGK1 gene (R=-0.8).

58

Results

Figure 12 Subgroup-specific DNA methylation patterns of SGK1 across medulloblastoma subgroups. Bisulfite conversion-based analysis of medulloblastoma samples (n=115) was performed using the Human Methylation 450K Array from Illumina. Methylation status of CpG sites in a 152 kb large region representing the SGK1 gene located on chromosome 6 (q23.2) is depicted. Tumors of the WNT and SHH- subgroup showed significantly more methylated (red) CpG sites of gene body and 3’UTR than tumors of Group 3 and Group 4 (R=-0.8). Unmethylated CpG sites are shown in blue. This experiment was performed by Prof. S. Pfister et al. in collaboration with Dr. D. Jones & Dr. M. Kool, DKFZ, Heidelberg.

3.1.5 Medulloblastoma Cell Lines Exhibit Different Variable SGK1 Expression Levels and Inducibility by Glucocorticoid Treatment In order to find an adequate cell culture model system for studying the biological role of SGK1 in medulloblastoma pathogenesis, various established medulloblastoma cell lines were investigated initially for endogenous SGK1 expression levels and inducibility. SGK1 mRNA levels were determined relative to the mean of two housekeeping genes (DCTN2, PGK1) in estabished cell lines DAOY, MED8A, ONS-76, D283 MED, UW228-2 and

59

Results

UW228-3 using QRT-PCR. Under basal conditions, SGK1 mRNA was overexpressed compared to normal cerebellum in DAOY and UW228 cell lines, whereas lower expression was found in MED8A, ONS-76, and D283 MED cell lines (Figure 13 A). The highest expression levels were observed in UW228 cell lines, likely due to gene-dosage effects caused by a focal high-level amplification at chromosome 6q23.2, harboring the SGK1 gene as assessed by array-CGH (data not shown). Furthermore, assessment of SGK1 abundance in these cell lines was also conducted on protein levels using Western blot analysis. To determine whether SGK1 is inducible on transcription level by administration of glucocorticoids using dexamethasone (DEX, 1 μM), cells were treated with DEX for 24h or left untreated (vehicle control). DEX was previously described to induce SGK1 expression on transcriptional levels via activation of the glucocorticoid receptor (GR)130. The upregulation of SGK1 mRNA levels in the presence of glucocorticoids (data not shown) was accompanied by a large increase of SGK1 protein levels in DAOY, ONS-76 and UW228 cell lines after 24h exposure to DEX, whereas no induction was observed in MED8A, and D283 MED cells (Figure 13 B). The endogenous SGK1 protein levels were also depicted and matched proportionately to the obtained mRNA expression levels shown in Figure 13 A. Taken together, these studies confirmed SGK1 being a downstream target of the glucocorticoid receptor in medulloblastoma cells.

Figure 13 A) Endogenous mRNA levels of SGK1 in various medulloblastoma cell lines. Expression values were given relative to the mean of two housekeeping genes (DCTN2, PGK1) and compared to normal cerebellum as measured by quantitative real-time PCR. B) Dexamethasone treatment of medullo- blastoma cell lines induces SGK1 protein expression. Upregulation of SGK1 protein levels in various medulloblastoma cell lines was detected by Western Blot analysis with or without dexamethasone (DEX, 1 μM) treatment for 24 h. Control cells were equally treated with vehicle only. SGK1 protein levels were strongly upregulated upon dexamethasone treatment in DAOY, ONS-76, and UW228 cells, whereas no induction was observed in MED8A, and D283 MED cells. These experiments were performed by D. Sturm, DKFZ, Heidelberg.

60

Results

3.2 FUNCTIONAL ANALYSES AFTER MODULATING SGK1 IN

MEDULLOBLATOMA CELL LINES

To get a better understanding of the functional role of SGK1 in medulloblastoma patho- genesis and course of disease, phenotypic assays were performed after modulating SGK1 expression in vitro. Since Daoy and UW228-3 medulloblastoma cell lines showed basic SGK1 expression levels detectable by QRT-PCR and Western blot analyses and moreover turned out to be inducible by dexamethasone, they were regarded as suitable cell culture models. Firstly, C-terminally HA-tagged SGK1 cDNA was cloned into a lentivirus compatible pLVX-vector to estabish stable SGK1 overexpression in cell lines. For lentivirus-mediated knockdown of SGK1, a commercially available set of shRNAs (short hairpin RNAs) targeting SGK1 and respective non- targeting shRNA (nt shRNA) control plasmids (pLKO.1 vector backbone) from Sigma were used. After producing infectious lentiviral particles containing either the SGK1 construct, or empty-vector / nt shRNA control particles, the (multiplicity of infection) MOI was calculated for each cell line. For both cell lines, an MOI of 5 was calculated, but satisfying phenotypic effects of SGK1 modulation were only seen with a transduction efficiency >90% using an MOI of 10. Therefore, this was maintained for all further experiments described below.

3.3 PHENOTYPIC ASSAYS AFTER STABLE OVEREXPRESSION OF SGK1 IN

MEDULLOBLASTOMA CELL LINES

A series of lentivirus-mediated overexpression experiments was performed to increase SGK1 expression levels. The readout included expression measurement of SGK1 on prorein level using Western blot analysis to confirm elevated SGK1 levels prior to investi- gation of resulting phenotype. Subsequently, assays evaluating proliferation, apoptosis and migration were conducted.

3.3.1 SGK1 Overexpression has no Effect on Proliferation For the purpose of demonstrating SGK1 specific effects on cell viability and growth rates, MTS assays were performed starting three days post transduction of Daoy and UW228-3 cell lines with lentiviruses containing the pLVX-constructs. After seeding the same numbers of SGK1-overexpressing cells and respective empty-vector control cells into 96- well plates, the readout was performed every 24h for a time series of five days. The cell

61

Results

viability was commensurated to the absorbance at 490nm. In both cell lines, Daoy and UW228-3, cells overexpressing SGK1 compared to control cells consistently did not significantly alter viability or growth characteristics (Figure 14).

Figure 14 Determination of cell viability and growth rates upon SGK1 overexpression. Depicted is the absorbance at 490nm representing the metabolic activity of Daoy (left panel) and UW228-3 (right panel) medulloblastoma cells using MTS-assay. Three days post transduction with respective pLVX-constructs MTS assays were performed every 24h for a total of five days. Cells overexressing SGK1 are shown as continuous lines, empty vector controls as dotted lines. The values are means ± SD of six technical replicates.

3.3.2 SGK1 Overexpression leads to Increased Migration Rates As a next step, the influence of SGK1 on medulloblastoma cell migration was investigated. Consequently, Daoy and UW228-3 cells were transduced with pLVX-SGK1- HA containing viruses or empty vector control viruses, respectively. Transduction occured four days prior to executing transwell assays by the usage of Boyden chambers filled with media containing 0.5% fetal bovine serum (FBS) in the upper compartment. Migration was directed towards a serum gradient achieved by a membrane with 8μm pore size separating the lower compartiment filled with media containing 10% FBS as attractant. Increase of migration propensity of cells harboring respective pLVX-constructs correlated exactly with their SGK1 protein expression levels (Figure 15 A, C, D). In parallel, MTS assays were performed to ensure equal viability and growth rates of the investigated medulloblastoma cells. As expected, no difference was seen in cell viability (Figure 15 B), whereas cells with elevated SGK1 protein levels showed a significant increase in migration rates up to 3-fold for Daoy and 2-fold for UW228-3 cells.

62

Results

Figure 15 Migration rates of medulloblastoma cells with or without overexpression of SGK1. Cells were either transduced with pLVX-SGK1-HA containing lentiviral particle or pLVX-empty vector control viruses (MOI 10) four days prior to the transwell assay. A) Five days post transduction, SGK1 protein expression levels in Daoy and UW228-3 cells were detected by Western Blot analysis. B) MTS assays of Daoy and UW228-3 cells were performed in parallel to the migration assay to ensure equal viability and growth rates of the cells. C) and D) Migration of transduced Daoy and UW228-3 cells occured from media containing 0.5% FBS in the upper compartment towards media with 10% FBS in the lower compartment using a 24-well format Boyden chamber with 8µm pore size. Increased migration rates of cells harboring respective pLVX-constructs correlated strongly with their SGK1 protein expression levels. The values are means ± SD of six technical replicates and normalized to empty vector control cells (p < 0.0001), respectively. These experiments were performed in collaboration with Prof. F. Lang et al. and Dr. Eva-Maria Schmidt, Institute of Physiology, Tübingen.

63

Results

3.3.3 SGK1 Overexpression has no Effect on Irradiation - but on Chemotherapy Resistance Current multimodal treatment protocols for standard- and high-risk medulloblastoma patients include surgical resection, different intensities of craniospinal gamma irradiation (17-36 Gy to the neuroaxis, 50-55 Gy to the posterior fossa), and varying regimens of chemotherapy101,102. As SGK1 was described to exhibit anti-apoptotic properties and to promote cell survival in several solid tumors125,171, the following approach was used to elucidate whether SGK1 protects against irradiation-induced cell death in medullo- blastoma: Daoy and UW228-3 cells were transduced with lentiviruses containing pLVX- constructs three days prior to irradiation at different doses. Viability and growth rates of medulloblastoma cells with or without overexpressing SGK1 were monitored up to three or five days, respectively, by using MTS assays. No differences of cells overexpressing SGK1 compared to control cells regarding radiation sensitivity were obtained with this experimental setup (Figure 16).

Figure 16 Viability and growth rates of medulloblastoma cells with or without overexpressing SGK1 exposed to irradiation at the indicated doses. Depicted is the absorbance at 490nm representing the metabolic activity of Daoy (left panel) and UW228-3 (right panel) medulloblastoma cells using MTS-assay. Three days post transduction with respective pLVX-constructs, cells were exposed to irradiation at the doses indicated and MTS readout was performed every 24h for a total of three days (A) or five days (B), respectively. Cells overexpressing SGK1 are shown as continuous lines, empty vector controls as dotted lines. The values are means ± SD of six technical replicates.

64

Results

To determine the role of SGK1 specifically in apoptosis-sensitivity towards radiotherapy, an additional experimental approach was choosen. Medulloblastoma cells were tested for the extent of apoptosis after irradiation with or without SGK1 overexpression by the usage of FACS-based Annexin V-PE/7AAD assay. This method exploits a property of most cells to translocate phosphatidylserine from the inner layer of the plasma membrane to the outer site, being detectable at the surface then. This exposure indicates the initial stages of apoptosis prior to morphological changes or loss of cellular integrity. Annexin V specifically binds to phosphatidylserine and can be used to measure apoptosis by covalently attaching a fluorochrome (e.g. phycoerythrin, PE). In order to identify late apoptotic or necrotic cells in addition, this measurement is complemented by the application of a DNA-intercalating fluorescent dye (7AAD, 7-aminoactinomycin), taking advantage of the disrupted membrane integrity in these cells. Daoy and UW228-3 medulloblastoma cells were transduced with pLVX-SGK1-HA or empty vector and three days later irradiated with 3 Gy and 6 Gy, whereas the control cells kept unirradiated. After another four days apoptotic cell populations were determined (early and late apoptosis) and plotted as percentages as depicted in Figure 17. In addition, the ratios of means were normalized to empty vector controls at respective irradiation doses which is showed as numbers at the bottom. This experiment confirmed the results measured by MTS assay, as no significant alterations in apoptosis rates due to SGK1 upregulation were obtained in both cell lines.

Figure 17 Determination of apoptosis rates in medulloblastoma cells with or without overexpressing SGK1 exposed to irradiation at the doses indicated. FACS-based apoptosis assay was performed on freshly harvested cells stained with Annexin V and 7-AAD detecting early and late apoptosis, respectively. Shown are percentages of apoptotic cell populations obtained from transduced Daoy (A) and UW228-3 (B) cells exposed to irradiation at different doses three days post transduction. Cells were either transduced with pLVX-SGK1-HA containing lentiviral particles or pLVX-empty vector control viruses (MOI 10) seven days prior to FACS analysis. The values are means ± SD of technical triplicates. The numbers at the bottom indicate ratios of means normalized to empty vector controls at different irradiation doses.

65

Results

Notably, beside of protection from apoptosis, also increased chemotherapy resistance upon glucocorticoid treatment has recently been described in various solid cancers including brain tumors174,198. Therefore, in vitro experiments were perfomed treating medulloblastoma cell lines with the alkylating agents 4-hydroperoxycyclophosphamide (4-HC) and lomustine (CCNU), which play a role among others in chemotherapy regimen for medulloblastoma patients. Both chemotherapeutics induce DNA doublestrand breaks (DSB). To evaluate the half-maximal effective concentration (EC50) of the substances after 48h in medulloblastoma cell lines, the effect of various concentrations applied on Daoy and UW228-3 wildtype cells was investigated by use of MTS assays. For 4-HC doses ranging from 0.5 μM to 40 μM and for CCNU doses ranging from 5 μM to 200 μM were tested. A significant reduction of tumor cell viability was observed in both cell lines in a dose-dependent manner. Dose response curves after 48h and EC50 values per cell line were indicated in Figure 18. UW228-3 cells turned out to be less sensitive for treatment than Daoy cells.

Figure 18 Dose response curves to determine the EC50 of 4-HC and CCNU after 48h exposure to medulloblastoma cell lines. The relative metabolic activity of Daoy (blue) and UW228-3 (red) medulloblastoma cells after 48h exposure to varying concentrations of 4-HC or CCNU as indicated, is shown using MTS-assay. The values are means ± SD of technical triplicates and plotted relative to control cells treated with vehicle alone. The half-maximal effective concentration (EC50) of each cell line was calculated as an indicator of response to the compound, respectively.

To elucidate whether SGK1 overexpression exhibits a protective effect on chemotherapy treatment, the same setup was tested in Daoy and UW228-3 cells with or without overxpressing SGK1. Subsequently, dose response curves for transduced medullo- blastoma cells were assessed, separately for either pLVX-SGK1-HA containing cells or empty-vector controls. Again, the EC50 was calculated as an indicator of response to the compound, respectively (Figure 19). After comparing the results within each cell line,

66

Results

SGK1 seems to play a role in mediation of resistance against 4-hydroperoxycyclo- phosphamide or lomustine in UW228-3 medulloblastoma cell lines. Minor effects were obtained for Daoy cells. These results still have to be validated in repeated experiments.

Figure 19 Dose response curves of medulloblastoma cells with or without overexpressing SGK1 exposed to 4-HC and CCNU for 48h, respectively. A-D) Depicted is the relative metabolic activity of Daoy (A and C) and UW228-3 (B and D) medulloblastoma cells transduced with respective pLVX-constructs after 48h exposure to varying concentrations of 4-HC or CCNU as indicated, using MTS-assay. The values are means ± SD of technical triplicates from a single experiment and plotted relative to control cells treated with vehicle alone. The half-maximal effective concentration (EC50) of each cell line with (red) or without (blue) overexpressing SGK1 was calculated as an indicator of response to the compound, respectively.

3.4 LENTIVIRUS-MEDIATED KNOCKDOWN OF SGK1 INDUCES APOPTOSIS IN

MEDULLOBLASTOMA CELL LINES

To determine the effects of the specific inhibition of SGK1 expression, lentivirus- mediated knockdown of SGK1 via RNA interference (RNAi) in medulloblastoma cell lines was performed. Daoy and UW228-3 cells were choosen as model based on their high endogenous SGK1 levels (mRNA and protein; Figure 13). Five days post transduction, decreased protein levels were measured. Using up to five different shRNAs targeting

67

Results

SGK1, knockdown efficiencies of 50 - 90% were achieved as assessed by Western blot analysis. Non-targeting shRNA was used as a control.

Figure 20 Apoptosis and cell cycle analysis of UW228-3 cells after efficient shRNA-mediated knockdown of SGK1. Western blot analysis stained for SGK1 and β-ACTIN (housekeeping protein) showing decreased SGK1 protein expression levels five days post transduction of UW228-3 cells with a set of five different shRNAs targeting SGK1 compared to non-targeting control (MOI 10). Seven days post transduction cell cycle was measured by FACS analysis as described by Nicoletti et al.195. Sub G1-fractions representing the percentage of apoptotic cell numbers were reproducibly higher in all SGK1-silenced cells. The values are means ± SD of three independent experiments.

Apoptosis and cell cycle analyses were performed seven days post transduction on cells stably expressing either one of the shRNAs against SGK1 or non-targeting shRNA. The percentages of extracted nuclei after propidium iodide staining (indicating DNA content) was measured by FACS analysis as described by Nicoletti et al.195. The assay revealed DNA content representing certain cell cycle phases (Sub G1, G1, S, G2/M) respectively. Apoptotic fractions (Sub G1) were significantly and reproducibly higher in all SGK1 silenced cells compared to controls (Figure 20 and Figure 21).

68

Results

Figure 21 Apoptosis and cell cycle analysis of Daoy cells after efficient shRNA-mediated knockdown of SGK1. Western blot analysis stained for SGK1 and β-ACTIN (housekeeping protein) showing decreased SGK1 protein expression levels five days post transduction of Daoy cells with a set of four different shRNAs targeting SGK1 compared to non-targeting control (MOI 10). Seven days post transduction cell cycle was measured by FACS analysis as described by Nicoletti et al.195 Sub G1-fractions representing the percentage of apoptotic cell numbers were higher in all SGK1-silenced cells. The values were obtained from a single experiment.

These results showing survival of Daoy and UW228-3 cells is dependent on SGK1 expression, raised the question whether SGK1 downregulation has a supportive effect on irradiation-induced apoptosis. Thus, stable knockdown of SGK1 was performed three days prior to irradiation at the doses indicated. Annexin V-PE/7AAD staining served as apoptosis assay, FACS measurement was conducted five days post transduction. Dose- dependent increase of irradiation-induced apoptosis was detected. In non-irradiated cells, the pure effect of efficient SGK1 knockdown on apoptosis rates is shown, confirming the results presented in Figure 20 and Figure 21. Enhanced apoptosis could be achieved by irradiation of SGK1-silenced cells. Regarding the calculated ratios of means normalized to non-targeting controls at different irradiation doses, the experiment revealed an additive effect on apoptosis induction concerning irradiation when combined with gene silencing of SGK1 (Figure 22). Consequently, no synergistic effect was observed.

69

Results

Figure 22 Determination of apoptosis rates in medulloblastoma cells after shRNA-mediated knockdown of SGK1 exposed to irradiation at the doses indicated. A-D) showing apoptosis assay on freshly harvested medulloblastoma cells stained with Annexin V and 7-AAD for early and late apoptosis, respectively. FACS analysis was performed five days post transduction with shRNAs targeting SGK1 and non-targeting control. ShRNA targeting BCL2 was used as a positive control for apoptosis induction. A) and C) represent apoptosis rates obtained from transduced Daoy and UW228-3 cells exposed to irradiation at different doses three days post transduction. The values are means ± SD of three independent experiments. The numbers at the bottom indicate ratios of means normalized to non-targeting controls at different irradiation doses. B) and D) showing exemplarily dot plot graphes of the FACS measurement.

70

Results

3.5 PHARMACOLOGICAL INHIBITION OF SGK1 BY A SMALL MOLECULE

(GSK650394) REDUCES CELL VIABILITY IN VITRO

To evaluate the role of SGK1 as a potential candidate for targeted drug therapy in medulloblastoma, the effect of administration of the SGK1 antagonist GSK650394 was tested in a panel of medullblastoma cell lines. This multi-kinase inhibitor had been developed as a prostate cancer therapeutic and was recently shown to effectively abrogate androgen-induced proliferation of prostate cancer cell lines. The selectivity of GSK650394 for SGK1 over that of Akt and other related kinases proved to be >30-fold, and it was >60-fold more selective for SGK1 over the upstream AGC kinase PDK1199.

Figure 23 Determination of apoptosis rates in various medulloblastoma cell lines after pharma- cological inhibition of SGK1. Cells were either treated with the SGK1 antagonist GSK650394 (10µM) or/and dexamethasone (1µM) or left untreated (vehicle alone) five days prior to FACS analysis. Αpoptosis assay was performed on freshly harvested cells stained with Annexin V and 7-AAD for early and late apoptosis, respectively. Shown are percentages of apoptotic cell populations. All medulloblastoma cells lines responded on GSK650394 exposure with an increase of apoptosis induction (grey bars). In Daoy and UW228-3 cells a partial rescue by dexamethasone could be achieved (black bars). The values are means ± SD of three independent experiments.

A significant pro-apoptotic impact of the compound on tumor cells upon SGK1 inhibition was observed in all investigated cell lines in a dose- and time-dependent manner. The half-maximal effective concentration (EC50) after a 5-day exposure was calculated to assess the response of individual cell lines to the administered substance (data not shown). EC50 values ranged from 4 μM in DAOY cells to 9 μM in the UW228 cell lines,

71

Results

being more resistant to the compound than all other cell lines tested likely due to harboring a high-level focal amplification of the SGK1 gene locus. For determination of apoptosis rates in various medulloblastoma cell lines after pharmacological inhibition of SGK1, cells were either treated with the SGK1 antagonist GSK650394 (10μM) with or without co-treatment with Dexamethasone (1μM) or left untreated (vehicle alone) five days prior to FACS analysis. All cell lines responded to GSK650394 exposure with an increase of apoptosis. In Daoy and UW228-3 cell lines, a partial rescue by dexamethasone could be achieved as indicated by the black bars in Figure 23.

3.6 NDRG1 IS A DIRECT DOWNSTREAM TARGET OF SGK1

Although various targets of SGK1 have been identified during the last years, the biological pathways altered by SGK1 signaling in medulloblastoma remain subject of further investigation. Murray et al. identified threonine residues in the n-myc downstream- regulated gene 1 protein (NDRG1-Thr346/356/366) that are phosphorylated by SGK1 but not by related kinases142. In order to explore SGK1 activity in medulloblastoma cells after lentivirus-mediated SGK1 modulation, phosphorylation status of NDRG1 was monitored using Western blot analysis. Accordingly, either specific knockdown of SGK1 using lentivirus-mediated RNAi or stable SGK1 overexpression was achieved in Daoy and UW228-3 cells. Non-targeting shRNA or empty vector were used as controls. Increase or decrease in phosphorylation levels should be a global effect in SGK1 silenced or overexpressing cells, respectively. Following Western blot analysis of total protein lysates, antibodies detecting SGK1, NDRG1, phospho-NDRG1 (Thr346) and β-ACTIN (loading control) were used to assess individual expression levels. The expression pattern of SGK1 changed according to the modulation, decrease upon efficient knockdown of SGK1 and strong upregulation upon overexpression, as expected and previously shown. Investigation of the phosphorylation site of NDRG1 at Thr346 revealed a decreased phosphorylation signal in SGK1 downregulated cells compared to non- targeting control (Figure 24 B). After overexpression of pLVX SGK1-HA, NDRG1 phosphorylation levels (Thr346) were highly elevated in comparison to empty vector control. Furthermore, the same pattern of down- or upregulation of SGK1 was observed in total NDRG1 protein levels. Taken together, NDRG1 is directly co-regulated with SGK1 on protein levels and constitutes a direct phosphorylation target of SGK1, indicating its activity in medulloblastoma cell lines (Figure 24 B).

72

Results

In addition, subgroup-specific gene expression of NDRG1 was determined in the same patient samples shown in Figure 11 A. This dataset (n=302) integrated different cohorts from the Heidelberg, Amsterdam, Paris and Cambridge series. Access was provided by the microarray analysis and visualization platform ’R2’ comprising untransformed, non- normalized NDRG1 gene expression values of medulloblastoma subgroups. In contrast to protein expression, NDRG1 was not co-regulated with SGK1 on mRNA level as indicated by comparing Figure 24 A with Figure 11 A.

Figure 24 A) Subgroup-specific gene expression pattern of NDRG1 in medulloblastoma. mRNA expression profiles were accessible via microarray analysis and visualization platform ‘R2’. Untransformed, non-normalized NDRG1 gene expression values of medulloblastoma samples (n=302), initially generated on Affymetrix gene expression arrays from different data sets were combined. Statistical significance was calculated using one-way ANOVA (p<0.0001). B) NDRG1 is a direct downstream target of SGK1. Western blot analysis showing decreased or elevated SGK1 protein expression levels five days post transduction of Daoy and UW228-3 cells after respective SGK1 modulation. Medulloblastoma cells were either transduced (MOI 10) with two different shRNAs targeting SGK1 to achieve an efficient knockdown of SGK1 or with pLVX-constructs for overexpressing SGK1. Staining for a phosphorylation-specific site of NDRG1 on Thr346 revealed a decreased phosphorylation signal in SGK1 downregulated cells compared to non-targeting control. After overexpression of pLVX SGK1-HA, NDRG1 phosphorylation on Thr346 was strongly enhanced in comparison to empty vector control. Moreover, the same pattern of down- or upregulation of SGK1 was observed in total NDRG1 protein levels.

3.7 EXPRESSION PROFILING: DEREGULATED GENES AFTER SGK1

KNOCKDOWN

In order to discover novel molecular functions of SGK1, which might complement the phenotypic results, genome wide mRNA expression analyses on Agilent microarrays (Whole Human Genome Microarray Kit, 4x44K, Agilent) were performed. To learn more about genes and pathways that are differentially regulated upon SGK1 knockdown in medulloblastoma cells, RNA of transduced Daoy and UW228-3 cells was isolated five days post transduction. The RNA obtained from cells containing either shRNA E8 or

73

Results

shRNA E9, both targeting SGK1, was labeled with a fluorophor and hybridized against labeled RNA of cells transduced with non-targeting shRNA on the array. After scanning the microrrays, the raw intensity data were preprocessed with the in-house platform ChipYard. The extracted list contained log2-ratios of normalized expression values. Annotation of oligonucleotide probes on gene expression arrays was retrieved via the PIMS© database (version: 1.26.1) by alignment of utilized sequences to EnsEMBL database version 60. Only unique valid oligonucleotides, meaning those which could be assigned to one definite gene, were included for the following analyses. Each list was sorted according to log2-ratios and the rank product over all experiments was calculated for each probe with the R-based software RankProd (http://bioinformatics.oxford- journals.org/content/22/22/2825.full).This approach modifies and extendes the rank product method proposed by Breitling et al.200. Using a percentage of false predictions (PFP) cutoff of 0.1 for the fold-change related rank product upon SGK1 knockdown, 172 genes were scored up- and 288 genes were downregulated (top 50 are shown in Supplementary Table 3 and Supplementary Table 4). To determine overrepresented pathways, the up- and downregulated candidates were examined for an over- representation of annotations with Gene Ontology terms, KEGG pathways and the entire set of canonical pathways from the Broad Institute’s Molecular Signatures Database (MSigDB v3.0; http://www.broadinstitute.org/gsea/msigdb/index.jsp). As a reference, all uniquely assigned possibly measurable genes from the arrays were used. Significant annotations were identified according to the relation of total genes versus deregulated genes in a pathway by Fisher’s exact test with Bonferroni’s correction method for multiple testing. Annotations with corrected p-values below or equal to 0.05 were considered to be significant. Using Gene Ontology biological process analysis, various enriched annotations were found for downregulated genes (n=288) upon SGK1 knockdown. Genes involved in e.g. blood vessel development, response to type I interferon and extracellular matrix organization were significantly enriched (Figure 25). Analysis of upregulated genes (n=172) after SGK1 knockdown revealed pathways involved in cell proliferation, developmental processes and phosphorylation of STAT5 protein (Table 3). Gene Ontology molecular function analysis identified genes playing a role for growth factor and activity, and organization of the extracellular matrix (Table 4). No significant enrichments were determined by usage of KEGG pathway analysis and MSig database.

74

Results

Figure 25 Enriched annotations of downregulated genes (n=288) after SGK1 knockdown using Gene Ontology biological process analysis. Number of downregulated genes in respective terms and corrected p-values are given, significant ones are shown in green. This bioinformatic analysis was performed in collaboration with Dr. M. Zapatka and Dr. R. Piro, DKFZ, Heidelberg.

75

Results

Table 3 Enriched annotations of upregulated genes (n=172) after SGK1 knockdown using Gene Ontology biological process analysis. This bioinformatic analysis was performed in collaboration with Dr. M. Zapatka and Dr. R. Piro, DKFZ, Heidelberg.

Annotation Number of Corrected GO term: Biological process genes p value GO:0042127 28 0.006 regulation of cell proliferation GO:0051239 33 0.02 regulation of multicellular organismal process GO:0051094 19 0.02 positive regulation of developmental process GO:0042522 4 0.03 regulation of tyrosine phosphorylation of Stat5 protein

Table 4 Enriched annotations of up- and downregulated genes (n=172; n=288) after SGK1 knockdown using Gene Ontology molecular function analysis. Upregulated genes are shown in red; downregulated genes are shown in green. This bioinformatic analysis was performed in collaboration with Dr. M. Zapatka and Dr. R. Piro, DKFZ, Heidelberg.

Annotation Up- and downregulated genes Corrected GO term: Molecular p value function GO:0070851 CSF2, FGF5, HBEGF, IL1A, IL1B, IL2, 0.02 growth factor receptor ITGB3 binding GO:0008083 CSF2, ESM1, FGF5, GDNF, HBEGF, 0.03 growth factor activity IL1B, IL2, NRG1 GO:0005125 CSF2, EDN1, IL1A, IL1B, IL2, IL8, 0.03 cytokine activity IL23A8, NRG1 GO:0005201 BGN, CHI3L1, COL11A1, COL12A1, 3,00E-05 extracellular matrix COL14A1, COL1A2, COL5A1, COL8A2, FBLN1, FN1, MGP

Furthermore, enrichment of total transcription factor binding affinities201,202 in a region of 1500 bp upstream to 500 bp downstream of the transcriptional start site (TSS) in the up- and downregulated candidates was investigated using a rank product score combined with an estimation of the false discovery rate (FDR) based on 10.000 random candidate sets. In doing so, binding sites for nine transcription factors were found to be significantly enriched in the downregulated gene set (n=288), whereas no significant enrichment was found in the upregulated genes (n=172), shown in Table 5.

76

Results

Table 5 Enrichment of total transcription factor binding affinities. A region of 1500 bp up-stream to 500 bp downstream of the transcriptional start site (TSS) in the downregulated candidates was investigated using a rank product score combined with an estimation of the false discovery rate (FDR) based on 10.000 random candidate sets. This bioinformatic analysis was performed in collaboration with Dr. M. Zapatka and Dr. R. Piro, DKFZ, Heidelberg.

Transcription factor False discovery rate (FDR) MZF1_1-4 0.002 TFAP2A 0.002 MZF1_5-13 0.026 Egr1 0.034 SP1 0.058 Mafb 0.059 REST 0.064 PLAG1 0.081 RREB1 0.076

The transcription factor ras responsive element binding protein 1 (RREB1) is co- expressed with SGK1 across medulloblastoma subgroups in expression profiling data sets (Figure 26; Figure 11A), whereas the other predicted transcription factors were not differentially expressed between molecular subgroups (Table 5; expression data not shown).

Figure 26 Subgroup-specific gene expression pattern of the transcription factor RREB1 in medulloblastoma. mRNA expression profiles were accessible via microarray analysis and visualization platform ‘R2’. Untransformed, non-normalized RREB1 gene expression values of medulloblastoma samples (n=302), initially generated on Affymetrix gene expression arrays from different data sets were combined. Statistical significance was calculated using one-way ANOVA (p<0.0001).

77

Discussion

4 DISCUSSION

Medulloblastoma is the most frequent malignant brain tumor occuring in childhood and constitutes one of the leading causes of childhood mortality. Current treatment decisions are based on histological tumor appearance, combined with clinical parameters such as patient age, presence of metastatic disease at primary diagnosis, and extend of surgical resection. Due to these factors, patients are stratified into clinical risk groups with distinct intensity of therapy (standard-/low-risk and high-risk). Multimodal therapy approaches include surgical resection, radiotherapy of the entire brain and spinal cord (in patients aged >3 years), followed by multi-agent chemotherapy. Therapeutic achievements of the last decades for patients suffering from medullo- blastoma have enabled 5-year overall survival (OS) rates of approximately 80% to 85% in children diagnosed with standard-risk disease nowadays104, and up to 70% 5-year OS in children classified with high-risk medulloblastoma105. Unfortunately, these remarkable improvement in clinical outcome is accompanied by marked quality of life issues, as many survivors experience significant intellectual and long-term neurological disabilities secondary to both disease and therapy106-109. Interestingly, the systematic use of empirically based cytotoxic treatment regimens has not been adapted to our increased understanding of medulloblastoma biology and development of novel therapies in recent years, yet. For novel treatment approaches, it is important to consider that medullo- blastoma comprises at least four distinct molecular subgroups with entirely different clinical and biological features84,85,110. There is strong evidence that clinical care of patients could be improved by the identification of new molecular biomarkers enabling appropriate stratification, and use of patient-specific, targeted anti-cancer agents87. Recent studies of retrospectively collected medulloblastoma samples have identified important molecular alterations and prognostic subgroups of this disease59,84- 86,88,99,110,111,203. After preliminary work demonstrating that DNA copy-number aberrations affecting chromosome 6q clearly define clinical and biological subgroups of medulloblastoma patients111, this study aimed at identifying candidate gene(s) targeted by these aberrations. This investigation was essential to get a better understanding of the underlying molecular pathomechanism, accounting for the astonishing differences in patient outcome according to chromosome 6q status.

78

Discussion

This thesis focused on SGK1, an attractive candidate gene on chromosome 6, which might potentially serve both as a prognostic marker and a novel therapeutic target in medulloblastoma.

4.1 SGK1 IS A PROGNOSTIC MARKER FOR HIGH-RISK-MEDULLOBLASTOMA

Integrative genome-wide analyses of DNA copy-number status, transcriptome profiles, and clinical follow-up data of 64 medulloblastoma samples revealed SGK1 as a promising candidate oncogene in medulloblastoma. SGK1 was identified based on its significant overexpression in tumors harboring a gain of chromosome 6q, compared to medulloblastomas with a balanced chromosome 6q, or monosomy 6. Positive correlation of SGK1 mRNA expression with chromosome 6q copy-number status was much stronger than expected by a simple gene-dosage effect (Figure 10), hinting at a functional impact of this kinase in medulloblastoma biology. Given that monosomy 6 is a genomic feature restricted to WNT medulloblastomas204, and gain of chromosome 6q seems be restricted to high-risk tumors (Group 3 and Group 4), SGK1 mRNA levels were further evaluated across the molecular medulloblastoma subgroups as suggested by Northcott et al.84. Subgroup-specific SGK1 expression patterns could be confirmed in our cohort as well as in three other independent data sets, demonstrating lowest expression levels in WNT and SHH tumors (associated with good/excellent prognosis), and overexpression in medulloblastomas allocated to Group 3 and Group 4 (characterized by poor prognosis) (Figure 11 A). Moreover, significantly higher SGK1 gene body and 3’UTR methylation was found on several CpG sites in the WNT and SHH-subgroups, whereas almost no methylation in the 3’ end of SGK1 was observed in Group 3 and Group 4 tumors (Figure 12). These results indicate a clear negative correlation of SGK1 mRNA expression levels and extent of methylation (R=-0.8), albeit the underlying mechanism of gene body and 3’UTR methylation still remains unclear. CpG sites are often grouped in clusters called CpG islands, which are predominantly present in the 5' regulatory regions of many genes. In several disease processes, such as cancer, gene promoter CpG islands acquire abnormal hypermethylation resulting in transcriptional silencing that can be inherited by daughter cells following cell division. Alterations in DNA methylation patterns, particulary associated with tumor suppressor genes, have been recognized as an important component in the development of cancer205-208. In contrast to gene silencing by hypermethylated promoter regions, gene- body methylation has been observed in Arabidopsis thaliana, where it is associated with

79

Discussion

active genes, therefore hypothesized to suppress spurious initiation of transcription within active genes209,210, and a similar function may exist in mammals. It has long been known that also in certain mammalian genes, methylation of gene bodies is positively correlated with elevated gene expression211,212. The underlying mechanism of SGK1 expression being negatively correlated with gene body methylation is not known yet. Hypermethylation of the gene body could influence the local chromatin structure to hinder transcription or alternatively, methyl CpG binding proteins, which are repressive for transcription, could be attracted213.

The strict correlation of SGK1 expression with prognosis renders abundant SGK1 expression a promising biomarker for high-risk medulloblastoma. The prognostic value of this novel finding was validated also on protein levels using an immunohistochemical approach. An independent large tumor cohort represented on a tissue microarray showed significant association of high SGK1 expression and poor overall survival (p=0.0168) (Figure 11 B). The application of an immunohistochemistry-based method to detect this novel biomarker confers the suitability for routine usage in a diagnostic neuropathological setting. While overexpression of SGK1 has previously been described in some hepatocellular carcinomas as well as in a high proportion of breast cancer specimens157-159,165, this is the first report providing evidence for its prognostic significance in tumors.

4.2 SGK1 IS A POTENTIAL DRUG TARGET IN MEDULLOBLASTOMA

The implication of SGK1 in tumorigenesis has previously been described in various solid tumors158-161. Several reports suggest a functional role of SGK1 in tumor biology by promoting cell survival and cell-cycle progression124,162.

4.2.1 Knockdown of SGK1 Induces Apoptosis in vitro Since abundant expression of SGK1 in high-risk medulloblastoma is associated with poor overall survival of patients in contrast to cases with low SGK1 expression and excellent prognosis (Figure 11), it was highly interesting to assess the potential functional role of SGK1 in medulloblastoma pathogenesis. Lentivirus-mediated knockdown of SGK1 in the medulloblastoma cell lines Daoy and UW228-3 dramatically induced apoptosis (Figure 20 and Figure 21). This phenotype points to an essential role

80

Discussion

for SGK1 in cell survival in medulloblastoma. The apoptosis rates could be increased in combination with irradiation three days post transduction in an additive, but not in a synergistic manner (Figure 22). Interestingly, as a close homologue to anti-apoptotic kinase AKT in both structure and function, SGK1 was described to function complementary to AKT in promoting cell survival by directly phosphorylating and inactivating the proapoptotic proteins forkhead transcrition factor FKHRL1 (FOXO3a) and BCL2-associated agonist of cell death (BAD)125,168. Survival factors, acting through kinases such as SGK1, AKT and PKA, induce endogenous BAD phosphorylation at three evolutionarily conserved sites (Ser- 112, Ser-136, and Ser-155) leading to loss of the ability of BAD to heterodimerize with the survival proteins BCL-XL or BCL-2. Phosphorylated BAD binds instead to 14-3-3 protein and is sequestered in the cytoplasm214,215. Can phosphorylation of BAD explain all of the apoptosis-inhibitory effects of SGK1? The answer is ‘probably not’, for various reasons. First, BAD is not a ubiquitously expressed protein, cell types that fail to express it are still protected by SGK1 or PKB/AKT216. Second, SGK1 phosphorylates several other proteins that are likely to regulate apoptosis, including e.g. FOXO3a, which is sequestered in the cytoplasm upon phosphorylation, resulting in an attenuation of its ability to transcribe pro-apoptotic genes such as Fas ligand214.

4.2.2 Overexpression of SGK1 Induces Chemotherapy Resistance and Increases Migration Rates in vitro Given the described pro-survial and cell cycle progression mediating effects of SGK1, phenotypic assays were also performed after stable SGK1 overexpression in the medulloblastoma cell lines Daoy and UW228-3. No effects on proliferation or inhibition of irradiation-induced apoptosis were detected in both cell lines (Figure 14, Figure 16, Figure 17), but in terms of resistance against alkylating agents interesting evidence was obtained. It seems that overexpression of SGK1 could have a protective role in response to 4-HC and CCNU in UW228-3 cells, because the EC50 value was increased in cells overexpressing SGK1 compared to their control counterparts, whereas only minor effects were detected in Daoy cells (Figure 19). This result has to be considered with caution, because the experiment was only performed once and it turned out that slightly less control cells were seeded than SGK1-overexpressing cells, which also could influence chemotherapy sensitivity. Hence, the result still has to be validated in future experiments. In addition, Daoy and UW228-3 medulloblastoma cells overexpressing SGK1 showed

81

Discussion

increased migration rates as compared to control cells (Figure 15 C, D). These phenotypes due to high SGK1 expression levels in vitro might also reflect the aggressive characteristics of high-risk medulloblastomas (Group 3 and Group 4) in vivo.

4.2.3 How Could the Obtained Phenotypes be Mediated by SGK1? To shed light on deregulated pathways and genes upon SGK1 knockdown, mRNA expression profiling was performed. Indeed, on transcriptome level only indirect effects could be detected. Microarray-based analysis revealed differentially expressed indirect targets after SGK1 knockdown, 172 genes were up- and 288 genes were downregulated (Top 50 are shown in Supplementary Table 3 and Supplementary Table 4). Gene Ontology annotations revealed significant enrichment for genes playing a role in blood vessel development, response to type I interferon, extracellular matrix organization, cell proliferation, developmental processes, phosphorylation of STAT5 protein, and growth factor- and cytokine activity (Figure 25, Table 3, Table 4). SGK1 seems to serve as an integration point for diverse signaling pathways. In fact, the obtained results provide a snapshot on deregulated pathways, which also could come up due to secondary effects. There are various mechanisms described in the literature, which could explain the obtained phenotypes upon SGK1 modulation in association with these deregulated pathways as discussed in the following section.

On transcriptional level, SGK1 has been shown to be upregulated upon dexamethasone treatment due to activation of the glucocorticoid receptor, mediated by a glucocorticoid response element (GRE) within the SGK1 promoter126,130. SGK1 protein levels are tightly balanced by ubiquitination and regulation of SGK1 activity is mediated by PI3K- signaling121. There are two conserved phosphorylation sites present in the activation loop of SGK1 and the hydrophobic domain at its C-terminus (Thr-256 and Ser-422), which both have to be phosphorylated for full kinase activity116-119. Lentivirus-mediated modulation of SGK1 was monitored on protein levels showing elevated SGK1 expression and moreover SGK1 activity was confirmed by increased phosphorylation of its downstream target NDRG1. Thus, protein expression and phosphorylation (Thr346) of NDRG1 are directly co-regulated with SGK1 in medulloblastoma cell lines (Figure 24 B).

NDRG1 was often described to play an ambiguous role in cancer-related processes. On one hand, NDRG1 is one of the most upregulated genes in hepatocellular carcinoma and

82

Discussion

high expression levels of NDRG1 were found to be significantly correlated with short overall survival, late tumor stage, large tumor size, the presence of vascular invasion, and high histological grade217. Furthermore, high NDRG1 expression is related to poor prognosis and angiogenesis in cervical adenocarcinoma and pancreatic cancer218,219. On the other hand, in other human malignancies such as gastric cancer, cervical cancer, and pancreatic cancers NDRG1 was suggested as a putative angiogenesis-suppressor gene218-220. Therefore, NDRG1 may promote or suppress tumor angiogenesis dependent on the cellular context221. For example, in human lung cancer cells, NDRG1 knockdown suppressed production of the potent angiogenic factors VEGF and CXCL-8, suggesting the involvement of reduced expression of such angiogenic factors in reduced angiogenesis resulting from NDRG1 knockdown222. Interestingly, the gene encoding cytokine CXCL-12 was downregulated upon SGK1 knockdown (Supplementary Table 4), was reported to act pro-metastatic via binding to the chemokine receptors CXCR-4 and CXCR-7223. It still remains unclear why NDRG1 could have a double-edged influence on cancer progression. NDRG1 is reported to be a multiphosphorylated protein224,225, while the role of phosphorylation is unknown but speculated to be related to the multitude of physiological functions of NDRG1222. Recent studies in glioblastoma cell lines revealed elevated NDRG1 phosphorylation levels in Temozolomide-resistant cell populations and NDRG1 overexpression was associated with poor overall survival in conventionally treated glioblastoma patients (unpublished data from Prof. W. Wick and J. Blaes, DKFZ, Heidelberg; personal communication). Therefore, NDRG1 could serve as a putative potential marker for chemotherapy resistance in glioblastoma. As shown above, NDRG1 and phosho-NDRG1 (Thr346) are co-regulated with SGK1 upon SGK1 modulation in medulloblastoma cell lines (Figure 24 B). In contrast, on mRNA level NDRG1 is not co-regulated with SGK1 (Figure 24 A). This discrepancy could indicate that regulation occurs post-translationally via SGK1, whereas the impact of NDRG1-phosphorylation by SGK1 in medulloblastoma still has to be unraveled. Thus, NDRG1 protein could play a crucial role in mediating the phenotypic effects and poor prognosis of SGK1 overexpression in vitro and in vivo, in addition to other signaling molecules, which are influenced by SGK1.

Another central player in SGK1 downstream signaling could be integrin beta 3 (ITGB3). ITGB3 was upregulated (log2 ratio 1.5) upon SGK1 knockdown compared to control cells (Supplementary Table 3). Growing evidence supports a central role for cooperative signaling between integrins, growth factor receptors and cytokine receptors in many

83

Discussion

aspects of tumor progression. This crosstalk not only regulates tumor cell adhesion, migration, invasion and survival, but also affects many aspects of the host response to cancer, in particular regarding the angiogenic endothelium226. Interestingly, another study on glioma cells revealed that the expression of pro-apoptotic ITGB3 was significantly downregulated in BCNU-resistant glioma cells and might be directly involved in the drug resistance of glioma cells. Subsequent dissection of signaling pathways denoted that extracellular signal-regulated kinase and unligated integrin-mediated cell death pathway may be involved in the pro-apoptotic role of ITGB3 in glioma cell death/survival and drug resistance227. Since BCNU is an alkylating drug, similar to CCNU, and ITGB3 was upregulated upon SGK1 knockdown, this might be another mechanism in mediating drug resistance in medulloblastoma. The ITGB3 expression status of medulloblastoma cells overexpressing SGK1 as well as the drug sensitivity of medulloblastoma cells after SGK1 knockdown needs to be further investigated.

Furthermore, integrins are crucial for cell migration and invasion228,229. They can bind to components of the extracellular matrix (ECM), thereby providing the traction that is necessary for cell motility and invasion and also remodelling of the ECM is controlled by integrins, which regulate the localization and activity of proteases230. Additionaly, deregulated integrin expression could also provide a reasonable mechanism for apoptosis-induction after depleting SGK1. Integrins are cell adhesion receptors that regulate cell survival and migration and can paradoxically initiate pro-survival as well as pro-apoptotic signals. Which pathway is more active depends on the ligation state of the respective surface integrins expressed by a given cell226. In adherent cells, in which many of the integrins are unligated, the unligated integrins initiate cleavage of caspase 8, triggering apoptosis through integrin-mediated death (IMD). At complete loss of adhesion, cell death is initiated through a process termed anoikis169,226. Consistent with this, a recent study showed that unligated integrin beta 3 (ITGB3) induced apoptosis in various solid tumors. Unligated integrin on the surface of adherent cells recruits and activates caspase 8, which in turn activates the proapoptotic factors Bid, Bax, and p53, and inhibits antiapoptotic Bcl-2. It was suggested that over-expression of ITGB3 may produce unligated ITGB3, which acts as negative modulator of cell survival, thereby leading to IMD-like cell death231. Notably, it has been previously shown, that activated SGK1 blocks apoptosis induced by loss of integrin-mediated cell attachment (anoikis)168.

84

Discussion

In this study, deregulated genes upon SGK1 knockdown were investigated on whether they share common transcription factor binding sites. Enrichment analysis revealed a significant overrepresentation of nine transcription factors likely to bind in regulatory regions of the candidate genes (Table 5). An interesting finding was, that ras responsive element binding protein 1 (RREB1) is co-expressed with SGK1 across medulloblastoma subgroups (Figure 26), whereas the other predicted transcription factors were not differentially expressed between subgroups. A possible explanation for this co-regulation with SGK1 in medulloblastoma is provided by the gene locus of RREB1 on 6p25, which could also be affected in monosomy or trisomy of chromosome 6 tumors, as it is the case for SGK1 on 6q23. Also a central role in SGK1 mediated signaling could be related to this transcription factor, as its effect became visible by the expression profiling experiment. RREB1 binds specifically to the RAS-responsive elements (RRE) of gene promoters, and activated Ras/Raf- signaling cascade prevents apoptosis in response to growth factors and mitogens232. Interstingly, a recent report demonstrated that knockdown of the transcription factor RREB1 inhibits collective cell migration in a scratch-wound healing assay of breast cancer cells, suppresses surface activity, and leads to the formation of immobile, tightly adherent cell colonies. Therefore RREB1 has been described to play an essential role in reducing cell-cell adhesion of epithelial cells within an interconnected cell cluster, which try to undergo dynamic changes in cell shape233. RREB1 is differentially expressed across medulloblastoma subgroups showing the same pattern as SGK1 in expression profiling data sets (Figure 26). High RREB1 levels correlate with Group 3 and Group 4 tumors which tend to metastasize and are associated with poor prognosis.

4.2.4 Small Molecule Inhibitor GSK650394 Induces Apoptosis in Medulloblastoma Cell Lines Supporting the observations in primary tumors, the obtained phenotypes after SGK1 modulation in vitro implicate a functional role of SGK1 in medulloblastoma pathogenesis, metastatic properties, and treatment response. These observations point to an eligibility exploiting SGK1 as promising drug target for the clinical management of medullo- blastoma. To test whether SGK1 function could be inhibited pharmacologically with respect to clinical application, various well-established medulloblastoma cell lines were treated with a small molecule SGK1 inhibitor (GSK650394). The therapeutic potential of SGK1 antagonist GSK650394 was previously evaluated in vitro and has been shown to

85

Discussion

effectively abrogate androgen-induced proliferation of prostate cancer cell lines199. The selectivity of GSK650394 for SGK1 over that of AKT and other related kinases proved to be >30-fold, and it was >60-fold more selective for SGK1 over the upstream AGC kinase PDK1199. A dramatic increase in apoptosis induction after exposing the cells for five days to the SGK1 antagonist GSK650394 (10μM) has been detected in all investigated medulloblastoma cell lines. Furthermore a partial rescue by co-administration of dexamethasone (1μM) could be achieved in Daoy and UW228-3 cell lines (Figure 23). Significant induction of SGK1 expression, accompanied by abundant SGK1 protein levels upon dexamethasone treatment was observed in well-established medullo- blastoma cell lines (Figure 13 B). Protection from apoptosis and increased chemotherapy resistance after glucocorticoid treatment has previously been described in a broad variety of solid cancers including brain tumors. It was shown that dexamethasone treatment inhibits cisplatin and 5- fluorouracil-induced apoptosis and promotes the growth of the malignant cells174. This phenomenon is associated with a glucocorticoid receptor-dependent, transcriptional induction of anti-apoptotic genes, including SGK1, which also protects breast cancer cells from growth factor starvation-induced apoptosis130,175. Nevertheless, glucocorticoids such as dexamethasone are clinically used in a supportive-care role for patients to effectively reduce peri-operative edema and furthermore to suppress nausea due to cancer chemotherapy or irradiation. However, no prospective clinical studies have assessed the effect of these steroids on the growth of solid tumors234-236. Since dexamethasone partially abrogated GSK650394 induced apoptosis (Figure 23), elevates SGK1 protein levels in medulloblastoma cell lines (Figure 13 B), and increases chemo- resistance after SGK1-overexpression (Figure 19), glucocorticoid-induced protection may counteract chemotherapy in solid tumors236,237. Consequently, to cirumvent therapy resistance in clinical practice, administration of glucocorticoids in medulloblastoma therapy should be avoided or substituted by other adjuvant substances whenever possible. Taken together, the presented findings indicating a prognostic role of SGK1 over- expression (Figure 11) combined with a more aggressive phenotype due to high SGK1 levels (Figure 15; Figure 19) and inducibility of apoptosis due to loss of SGK1 function (Figure 20; Figure 21; Figure 23) provide strong evidence for SGK1 as novel therapeutic target in high-risk medulloblastomas. This specified therapeutic approach might support or even modify conventional therapy regimens to improve individual patient outcome and diminish side effects of conventional cytotoxic chemotherapy.

86

Conclusions and Outlook

5 CONCLUSIONS AND OUTLOOK

Previous data showing that chromosome 6q status defines clinical subgroups for the outcome of medulloblastoma patients constituted the starting point of the molecular analyses performed in the present study. Global gene expression profiling and immunohistochemical examination of medulloblastoma samples identified SGK1 at chromosome band 6q23 as a novel prognostic marker and thus interesting candidate gene on chromosome 6. Functional analyses in vitro demonstrated that depletion of SGK1 function induces apoptosis, whereas ectopic overexpression of SGK1 results in increased migration. The obtained phenotypes after SGK1 modulation in vitro likely result from an interplay between diverse deregulated pathways including extracellular matrix organization, cell proliferation, developmental processes, blood vessel development, as well as growth factor- and cytokine activity. Despite the exact mechanisms still need to be unraveled, the findings provide strong in vitro evidence for the pivotal role of SGK1 in the pathogenesis of medulloblastoma. This highlights the importance of SGK1 as a valuable therapeutic target. At the same time, SGK1-overexpressing cells constitute a convenient in vitro model for high-risk medulloblastoma showing copy-number gain of chromosome 6q and consecutive overexpression of SGK1, which offers the possibility for pre-clinical evaluation of novel therapy options using pharmacological SGK1 inhibition or other more indirect ways of inhibiting the same pathway. Consequently, a mouse model for studying the impact of SGK1 in high-risk medulloblastoma in terms of microenvironment, angiogenesis, metastatic dissemination, and treatment response would be highly interesting. Given that high SGK1 expression is a frequent phenomenon in Group 3 and Group 4 tumors associated with poor prognosis, SGK1 comprises a potential drug target for affected medulloblastoma patients. Finally, a targeted therapy approach, adjusted to tumor biology, might open excellent prospects towards personalizing medulloblastoma therapy to improve individual outcome in the future.

87

References

6 REFERENCES

1 http://www.cancerhelp.cancerresearchuk.org. (2012). 2 Weinberg, R. A. The biology of cancer. (Garland Science, 2007). 3 Hanahan, D. & Weinberg, R. A. The hallmarks of cancer. Cell 100, 57-70, (2000). 4 Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: the next generation. Cell 144, 646-674, (2011). 5 Downward, J. Targeting RAS signalling pathways in cancer therapy. Nat Rev Cancer 3, 11-22, (2003). 6 Blobe, G. C., Schiemann, W. P. & Lodish, H. F. Role of transforming growth factor beta in human disease. N Engl J Med 342, 1350-1358, (2000). 7 Alberts. Molecular Biology of the Cell. Vol. 5th ed (Garland Science, 2008). 8 Levine, A. J. p53, the cellular gatekeeper for growth and division. Cell 88, 323- 331, (1997). 9 White, E. & DiPaola, R. S. The double-edged sword of autophagy modulation in cancer. Clin Cancer Res 15, 5308-5316, (2009). 10 Hayflick, L. The strategy of senescence. Gerontologist 14, 37-45, (1974). 11 Cesare, A. J. & Reddel, R. R. Alternative lengthening of telomeres: models, mechanisms and implications. Nat Rev Genet 11, 319-330, (2010). 12 Folkman, J. Role of angiogenesis in tumor growth and metastasis. Semin Oncol 29, 15-18, (2002). 13 Bergers, G. & Benjamin, L. E. Tumorigenesis and the angiogenic switch. Nat Rev Cancer 3, 401-410, (2003). 14 Chiang, A. C. & Massague, J. Molecular basis of metastasis. N Engl J Med 359, 2814-2823, (2008). 15 Chambers, A. F., Groom, A. C. & MacDonald, I. C. Dissemination and growth of cancer cells in metastatic sites. Nat Rev Cancer 2, 563-572, (2002). 16 Egeblad, M. & Werb, Z. New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer 2, 161-174, (2002). 17 Takeda, K. et al. Critical role for tumor necrosis factor-related apoptosis-inducing ligand in immune surveillance against tumor development. J Exp Med 195, 161- 169, (2002). 18 Warburg, O. On respiratory impairment in cancer cells. Science 124, 269-270, (1956). 19 Vander Heiden, M. G., Cantley, L. C. & Thompson, C. B. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 324, 1029-1033, (2009). 20 Younes, M., Ertan, A., Lechago, L. V., Somoano, J. & Lechago, J. Human erythrocyte glucose transporter (Glut1) is immunohistochemically detected as a late event during malignant progression in Barrett's metaplasia. Cancer Epidemiol Biomarkers Prev 6, 303-305, (1997). 21 Macheda, M. L., Rogers, S. & Best, J. D. Molecular and cellular regulation of glucose transporter (GLUT) proteins in cancer. J Cell Physiol 202, 654-662, (2005). 22 Artandi, S. E. & DePinho, R. A. Telomeres and telomerase in cancer. Carcinogenesis 31, 9-18, (2010). 23 Negrini, S., Gorgoulis, V. G. & Halazonetis, T. D. Genomic instability--an evolving hallmark of cancer. Nat Rev Mol Cell Biol 11, 220-228, (2010).

88

References

24 Jackson, S. P. The DNA-damage response: new molecular insights and new approaches to cancer therapy. Biochem Soc Trans 37, 483-494, (2009). 25 Grivennikov, S. I., Greten, F. R. & Karin, M. Immunity, inflammation, and cancer. Cell 140, 883-899, (2010). 26 Vogelstein, B. et al. Genetic alterations during colorectal-tumor development. N Engl J Med 319, 525-532, (1988). 27 Vogelstein, B. & Kinzler, K. W. The multistep nature of cancer. Trends Genet 9, 138-141, (1993). 28 Hudson, T. J. et al. International network of cancer genome projects. Nature 464, 993-998, (2010). 29 Croce, C. M. Oncogenes and cancer. N Engl J Med 358, 502-511, (2008). 30 Stehelin, D., Varmus, H. E., Bishop, J. M. & Vogt, P. K. DNA related to the transforming gene(s) of avian sarcoma viruses is present in normal avian DNA. Nature 260, 170-173, (1976). 31 Yamamoto, F. & Perucho, M. Activation of a human c-K-ras oncogene. Nucleic Acids Res 12, 8873-8885, (1984). 32 Weiss, R. The myc oncogene in man and birds. Nature 299, 9-10, (1982). 33 Forrester, K., Almoguera, C., Han, K., Grizzle, W. E. & Perucho, M. Detection of high incidence of K-ras oncogenes during human colon tumorigenesis. Nature 327, 298-303, (1987). 34 Marshall, M. S. Ras target proteins in eukaryotic cells. Faseb J 9, 1311-1318, (1995). 35 Kinzler, K. W. & Vogelstein, B. Lessons from hereditary colorectal cancer. Cell 87, 159-170, (1996). 36 Weinstein, I. B. & Joe, A. K. Mechanisms of disease: Oncogene addiction--a rationale for molecular targeting in cancer therapy. Nat Clin Pract Oncol 3, 448- 457, (2006). 37 Knudson, A. G., Jr. Mutation and cancer: statistical study of retinoblastoma. Proc Natl Acad Sci U S A 68, 820-823, (1971). 38 Lees, J. A. et al. The retinoblastoma protein binds to a family of E2F transcription factors. Mol Cell Biol 13, 7813-7825, (1993). 39 Weinberg, R. A. The retinoblastoma protein and cell cycle control. Cell 81, 323- 330, (1995). 40 Burkhart, D. L. & Sage, J. Cellular mechanisms of tumour suppression by the retinoblastoma gene. Nat Rev Cancer 8, 671-682, (2008). 41 Osaki, M., Oshimura, M. & Ito, H. PI3K-Akt pathway: its functions and alterations in human cancer. Apoptosis 9, 667-676, (2004). 42 Baker, S. J., Markowitz, S., Fearon, E. R., Willson, J. K. & Vogelstein, B. Suppression of human colorectal carcinoma cell growth by wild-type p53. Science 249, 912-915, (1990). 43 Meek, D. W. Tumour suppression by p53: a role for the DNA damage response? Nat Rev Cancer 9, 714-723, (2009). 44 Milner, J. & Medcalf, E. A. Cotranslation of activated mutant p53 with wild type drives the wild-type p53 protein into the mutant conformation. Cell 65, 765-774, (1991). 45 Prives, C. Signaling to p53: breaking the MDM2-p53 circuit. Cell 95, 5-8, (1998). 46 Zurawel, R. H., Allen, C., Wechsler-Reya, R., Scott, M. P. & Raffel, C. Evidence that haploinsufficiency of Ptch leads to medulloblastoma in mice. Genes Cancer 28, 77-81, (2000). 47 Fero, M. L., Randel, E., Gurley, K. E., Roberts, J. M. & Kemp, C. J. The murine gene p27Kip1 is haplo-insufficient for tumour suppression. Nature 396, 177-180, (1998).

89

References

48 Vogelstein, B. & Kinzler, K. W. Cancer genes and the pathways they control. Nat Med 10, 789-799, (2004). 49 Fong, P. C. et al. Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. N Engl J Med 361, 123-134, (2009). 50 Louis, D. N. et al. The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol 114, 97-109, (2007). 51 Hoffman, S., Propp, J. M. & McCarthy, B. J. Temporal trends in incidence of primary brain tumors in the United States, 1985-1999. Neuro Oncol 8, 27-37, (2006). 52 Ellison, D. Classifying the medulloblastoma: insights from morphology and molecular genetics. Neuropathol Appl Neurobiol 28, 257-282, (2002). 53 Ellison, D. W. Childhood medulloblastoma: novel approaches to the classification of a heterogeneous disease. Acta Neuropathol 120, 305-316, (2010). 54 McManamy, C. S. et al. Nodule formation and desmoplasia in medulloblastomas- defining the nodular/desmoplastic variant and its biological behavior. Brain Pathol 17, 151-164, (2007). 55 Rodini, C. O. et al. Aberrant signaling pathways in medulloblastomas: a stem cell connection. Arq Neuropsiquiatr 68, 947-952, (2010). 56 Yang, Z. J. et al. Medulloblastoma can be initiated by deletion of Patched in lineage-restricted progenitors or stem cells. Cancer Cell 14, 135-145, (2008). 57 Eberhart, C. G. In search of the medulloblast: neural stem cells and embryonal brain tumors. Neurosurg Clin N Am 18, 59-69, viii-ix, (2007). 58 Eberhart, C. G. et al. Comparative genomic hybridization detects an increased number of chromosomal alterations in large cell/anaplastic medulloblastomas. Brain Pathol 12, 36-44, (2002). 59 Korshunov, A. et al. Accumulation of genomic aberrations during clinical progression of medulloblastoma. Acta Neuropathol 116, 383-390, (2008). 60 Lindberg, E. et al. Concurrent gain of 17q and the MYC oncogene in a medullomyoblastoma. Neuropathology 27, 556-560, (2007). 61 Hatten, M. E. & Roussel, M. F. Development and cancer of the cerebellum. Trends Neurosci 34, 134-142, (2011). 62 Fan, X. & Eberhart, C. G. Medulloblastoma stem cells. J Clin Oncol 26, 2821- 2827, (2008). 63 Rausch, T. et al. Genome sequencing of pediatric medulloblastoma links catastrophic DNA rearrangements with TP53 mutations. Cell 148, 59-71, (2012). 64 Goodrich, L. V. & Scott, M. P. Hedgehog and patched in neural development and disease. Neuron 21, 1243-1257, (1998). 65 Kimonis, V. E. et al. Clinical manifestations in 105 persons with nevoid basal cell carcinoma syndrome. Am J Med Genet 69, 299-308, (1997). 66 Pietsch, T. et al. Medulloblastomas of the desmoplastic variant carry mutations of the human homologue of Drosophila patched. Cancer Res 57, 2085-2088, (1997). 67 Raffel, C. et al. Sporadic medulloblastomas contain PTCH mutations. Cancer Res 57, 842-845, (1997). 68 Reifenberger, J. et al. Missense mutations in SMOH in sporadic basal cell carcinomas of the skin and primitive neuroectodermal tumors of the central nervous system. Cancer Res 58, 1798-1803, (1998). 69 Taylor, M. D. et al. Mutations in SUFU predispose to medulloblastoma. Nat Genet 31, 306-310, (2002). 70 Wechsler-Reya, R. & Scott, M. P. The developmental biology of brain tumors. Annu Rev Neurosci 24, 385-428, (2001).

90

References

71 Romer, J. & Curran, T. Targeting medulloblastoma: small-molecule inhibitors of the Sonic Hedgehog pathway as potential cancer therapeutics. Cancer Res 65, 4975-4978, (2005). 72 Malkin, D. et al. Germ line p53 mutations in a familial syndrome of breast cancer, sarcomas, and other neoplasms. Science 250, 1233-1238, (1990). 73 Li, F. P. & Fraumeni, J. F., Jr. Soft-tissue sarcomas, breast cancer, and other neoplasms. A familial syndrome? Ann Intern Med 71, 747-752, (1969). 74 Forbes, S. A. et al. COSMIC: mining complete cancer genomes in the Catalogue of Somatic Mutations in Cancer. Nucleic Acids Res 39, D945-950, (2011). 75 Vogelstein, B., Lane, D. & Levine, A. J. Surfing the p53 network. Nature 408, 307- 310, (2000). 76 Turcot, J., Despres, J. P. & St Pierre, F. Malignant tumors of the central nervous system associated with familial polyposis of the colon: report of two cases. Dis Colon Rectum 2, 465-468, (1959). 77 Mori, T. et al. Germ-line and somatic mutations of the APC gene in patients with Turcot syndrome and analysis of APC mutations in brain tumors. Genes Chromosomes Cancer 9, 168-172, (1994). 78 Rubinfeld, B., Albert, I., Porfiri, E., Munemitsu, S. & Polakis, P. Loss of beta- catenin regulation by the APC tumor suppressor protein correlates with loss of structure due to common somatic mutations of the gene. Cancer Res 57, 4624- 4630, (1997). 79 Iwao, K. et al. Activation of the beta-catenin gene by interstitial deletions involving exon 3 in primary colorectal carcinomas without adenomatous polyposis coli mutations. Cancer Res 58, 1021-1026, (1998). 80 Tetsu, O. & McCormick, F. Beta-catenin regulates expression of cyclin D1 in colon carcinoma cells. Nature 398, 422-426, (1999). 81 He, T. C. et al. Identification of c-MYC as a target of the APC pathway. Science 281, 1509-1512, (1998). 82 Pomeroy, S. L. et al. Prediction of central nervous system embryonal tumour outcome based on gene expression. Nature 415, 436-442, (2002). 83 Taylor, M. D. et al. Molecular subgroups of medulloblastoma: the current consensus. Acta Neuropathol 123, 465-472, (2012). 84 Northcott, P. A. et al. Medulloblastoma Comprises Four Distinct Molecular Variants. J Clin Oncol, (2010). 85 Kool, M. et al. Integrated genomics identifies five medulloblastoma subtypes with distinct genetic profiles, pathway signatures and clinicopathological features. PLoS One 3, e3088, (2008). 86 Cho, Y. J. et al. Integrative Genomic Analysis of Medulloblastoma Identifies a Molecular Subgroup That Drives Poor Clinical Outcome. J Clin Oncol, (2010). 87 Northcott, P. A., Korshunov, A., Pfister, S. M. & Taylor, M. D. The clinical implications of medulloblastoma subgroups. Nat Rev Neurol, (2012). 88 Korshunov, A. et al. Adult and pediatric medulloblastomas are genetically distinct and require different algorithms for molecular risk stratification. J Clin Oncol 28, 3054-3060, (2010). 89 Koch, A. et al. Mutations of the Wnt antagonist AXIN2 (Conductin) result in TCF- dependent transcription in medulloblastomas. Int J Cancer 121, 284-291, (2007). 90 Ellison, D. W. et al. beta-Catenin status predicts a favorable outcome in childhood medulloblastoma: the United Kingdom Children's Cancer Study Group Brain Tumour Committee. J Clin Oncol 23, 7951-7957, (2005). 91 Eberhart, C. G., Tihan, T. & Burger, P. C. Nuclear localization and mutation of beta-catenin in medulloblastomas. J Neuropathol Exp Neurol 59, 333-337, (2000).

91

References

92 Gibson, P. et al. Subtypes of medulloblastoma have distinct developmental origins. Nature 468, 1095-1099, (2010). 93 Goodrich, L. V., Milenkovic, L., Higgins, K. M. & Scott, M. P. Altered neural cell fates and medulloblastoma in mouse patched mutants. Science 277, 1109-1113, (1997). 94 Lee, Y. et al. A molecular fingerprint for medulloblastoma. Cancer Res 63, 5428- 5437, (2003). 95 Northcott, P. A. et al. Pediatric and adult sonic hedgehog medulloblastomas are clinically and molecularly distinct. Acta Neuropathol 122, 231-240, (2011). 96 Rudin, C. M. et al. Treatment of medulloblastoma with hedgehog pathway inhibitor GDC-0449. N Engl J Med 361, 1173-1178, (2009). 97 Pei, Y. et al. An animal model of MYC-driven medulloblastoma. Cancer Cell 21, 155-167, (2012). 98 Kawauchi, D. et al. A mouse model of the most aggressive subgroup of human medulloblastoma. Cancer Cell 21, 168-180, (2012). 99 Remke, M. et al. Adult medulloblastoma comprises three major molecular variants. J Clin Oncol 29, 2717-2723, (2011). 100 Kool, M. et al. Molecular subgroups of medulloblastoma: an international meta- analysis of transcriptome, genetic aberrations, and clinical data of WNT, SHH, Group 3, and Group 4 medulloblastomas. Acta Neuropathol 123, 473-484, (2012). 101 Lafay-Cousin, L. & Strother, D. Current treatment approaches for infants with malignant central nervous system tumors. Oncologist 14, 433-444, (2009). 102 Kortmann, R. D. et al. Postoperative neoadjuvant chemotherapy before radiotherapy as compared to immediate radiotherapy followed by maintenance chemotherapy in the treatment of medulloblastoma in childhood: results of the German prospective randomized trial HIT '91. Int J Radiat Oncol Biol Phys 46, 269-279, (2000). 103 http://www.neurosurgery.tv/medulloblastoma.html. 104 Packer, R. J. et al. Phase III study of craniospinal radiation therapy followed by adjuvant chemotherapy for newly diagnosed average-risk medulloblastoma. J Clin Oncol 24, 4202-4208, (2006). 105 Gajjar, A. et al. Risk-adapted craniospinal radiotherapy followed by high-dose chemotherapy and stem-cell rescue in children with newly diagnosed medulloblastoma (St Jude Medulloblastoma-96): long-term results from a prospective, multicentre trial. Lancet Oncol 7, 813-820, (2006). 106 Hoppe-Hirsch, E. et al. Medulloblastoma in childhood: progressive intellectual deterioration. Childs Nerv Syst 6, 60-65, (1990). 107 Mulhern, R. K., Merchant, T. E., Gajjar, A., Reddick, W. E. & Kun, L. E. Late neurocognitive sequelae in survivors of brain tumours in childhood. Lancet Oncol 5, 399-408, (2004). 108 Silber, J. H. et al. Whole-brain irradiation and decline in intelligence: the influence of dose and age on IQ score. J Clin Oncol 10, 1390-1396, (1992). 109 Laughton, S. J. et al. Endocrine outcomes for children with embryonal brain tumors after risk-adapted craniospinal and conformal primary-site irradiation and high-dose chemotherapy with stem-cell rescue on the SJMB-96 trial. J Clin Oncol 26, 1112-1118, (2008). 110 Thompson, M. C. et al. Genomics identifies medulloblastoma subgroups that are enriched for specific genetic alterations. J Clin Oncol 24, 1924-1931, (2006). 111 Pfister, S. et al. Outcome prediction in pediatric medulloblastoma based on DNA copy-number aberrations of chromosomes 6q and 17q and the MYC and MYCN loci. J Clin Oncol 27, 1627-1636, (2009).

92

References

112 Parker, P. J. & Parkinson, S. J. AGC protein kinase phosphorylation and protein kinase C. Biochem Soc Trans 29, 860-863, (2001). 113 Frodin, M. et al. A phosphoserine/threonine-binding pocket in AGC kinases and PDK1 mediates activation by hydrophobic motif phosphorylation. Embo J 21, 5396-5407, (2002). 114 Webster, M. K., Goya, L., Ge, Y., Maiyar, A. C. & Firestone, G. L. Characterization of , a novel member of the serine/threonine protein kinase gene family which is transcriptionally induced by glucocorticoids and serum. Mol Cell Biol 13, 2031-2040, (1993). 115 Lang, F. & Cohen, P. Regulation and physiological roles of serum- and glucocorticoid-induced protein kinase isoforms. Sci STKE 2001, re17, (2001). 116 Alessi, D. R., Kozlowski, M. T., Weng, Q. P., Morrice, N. & Avruch, J. 3- Phosphoinositide-dependent protein kinase 1 (PDK1) phosphorylates and activates the p70 S6 kinase in vivo and in vitro. Curr Biol 8, 69-81, (1998). 117 Le Good, J. A. et al. Protein kinase C isotypes controlled by phosphoinositide 3- kinase through the protein kinase PDK1. Science 281, 2042-2045, (1998). 118 Vanhaesebroeck, B. & Alessi, D. R. The PI3K-PDK1 connection: more than just a road to PKB. Biochem J 346 Pt 3, 561-576, (2000). 119 Pullen, N. et al. Phosphorylation and activation of p70s6k by PDK1. Science 279, 707-710, (1998). 120 Kobayashi, T. & Cohen, P. Activation of serum- and glucocorticoid-regulated protein kinase by agonists that activate phosphatidylinositide 3-kinase is mediated by 3-phosphoinositide-dependent protein kinase-1 (PDK1) and PDK2. Biochem J 339 ( Pt 2), 319-328, (1999). 121 Park, J. et al. Serum and glucocorticoid-inducible kinase (SGK) is a target of the PI 3-kinase-stimulated signaling pathway. Embo J 18, 3024-3033, (1999). 122 http://mutagenetix.utsouthwestern.edu. 123 Waldegger, S., Barth, P., Raber, G. & Lang, F. Cloning and characterization of a putative human serine/threonine protein kinase transcriptionally modified during anisotonic and isotonic alterations of cell volume. Proc Natl Acad Sci U S A 94, 4440-4445, (1997). 124 Buse, P. et al. Cell cycle and hormonal control of nuclear-cytoplasmic localization of the serum- and glucocorticoid-inducible protein kinase, Sgk, in mammary tumor cells. A novel convergence point of anti-proliferative and proliferative cell signaling pathways. J Biol Chem 274, 7253-7263, (1999). 125 Brunet, A. et al. Protein kinase SGK mediates survival signals by phosphorylating the forkhead transcription factor FKHRL1 (FOXO3a). Mol Cell Biol 21, 952-965, (2001). 126 Maiyar, A. C., Phu, P. T., Huang, A. J. & Firestone, G. L. Repression of glucocorticoid receptor transactivation and DNA binding of a glucocorticoid response element within the serum/glucocorticoid-inducible protein kinase (sgk) gene promoter by the p53 tumor suppressor protein. Mol Endocrinol 11, 312-329, (1997). 127 Pearce, L. R., Komander, D. & Alessi, D. R. The nuts and bolts of AGC protein kinases. Nat Rev Mol Cell Biol 11, 9-22, (2010). 128 Hunter, T. Protein kinases and phosphatases: the yin and yang of protein phosphorylation and signaling. Cell 80, 225-236, (1995). 129 Brickley, D. R., Mikosz, C. A., Hagan, C. R. & Conzen, S. D. Ubiquitin modification of serum and glucocorticoid-induced protein kinase-1 (SGK-1). J Biol Chem 277, 43064-43070, (2002). 130 Mikosz, C. A., Brickley, D. R., Sharkey, M. S., Moran, T. W. & Conzen, S. D. Glucocorticoid receptor-mediated protection from apoptosis is associated with

93

References

induction of the serine/threonine survival kinase gene, sgk-1. J Biol Chem 276, 16649-16654, (2001). 131 Naray-Fejes-Toth, A., Fejes-Toth, G., Volk, K. A. & Stokes, J. B. SGK is a primary glucocorticoid-induced gene in the human. J Steroid Biochem Mol Biol 75, 51-56, (2000). 132 Itani, S. I., Zhou, Q., Pories, W. J., MacDonald, K. G. & Dohm, G. L. Involvement of protein kinase C in human skeletal muscle insulin resistance and obesity. Diabetes 49, 1353-1358, (2000). 133 Alliston, T. N., Maiyar, A. C., Buse, P., Firestone, G. L. & Richards, J. S. Follicle stimulating hormone-regulated expression of serum/glucocorticoid-inducible kinase in rat ovarian granulosa cells: a functional role for the Sp1 family in promoter activity. Mol Endocrinol 11, 1934-1949, (1997). 134 Alliston, T. N., Gonzalez-Robayna, I. J., Buse, P., Firestone, G. L. & Richards, J. S. Expression and localization of serum/glucocorticoid-induced kinase in the rat ovary: relation to follicular growth and differentiation. Endocrinology 141, 385- 395, (2000). 135 Waldegger, S. et al. h-sgk serine-threonine protein kinase gene as transcriptional target of transforming growth factor beta in human intestine. Gastroenterology 116, 1081-1088, (1999). 136 Cowling, R. T. & Birnboim, H. C. Expression of serum- and glucocorticoid- regulated kinase (sgk) mRNA is up-regulated by GM-CSF and other proinflammatory mediators in human granulocytes. J Leukoc Biol 67, 240-248, (2000). 137 Mizuno, H. & Nishida, E. The ERK MAP kinase pathway mediates induction of SGK (serum- and glucocorticoid-inducible kinase) by growth factors. Genes Cells 6, 261-268, (2001). 138 Leong, M. L., Maiyar, A. C., Kim, B., O'Keeffe, B. A. & Firestone, G. L. Expression of the serum- and glucocorticoid-inducible protein kinase, Sgk, is a cell survival response to multiple types of environmental stress stimuli in mammary epithelial cells. J Biol Chem 278, 5871-5882, (2003). 139 Bogusz, A. M., Brickley, D. R., Pew, T. & Conzen, S. D. A novel N-terminal hydrophobic motif mediates constitutive degradation of serum- and glucocorticoid-induced kinase-1 by the ubiquitin-proteasome pathway. Febs J 273, 2913-2928, (2006). 140 Zhou, R. & Snyder, P. M. Nedd4-2 phosphorylation induces serum and glucocorticoid-regulated kinase (SGK) ubiquitination and degradation. J Biol Chem 280, 4518-4523, (2005). 141 Raikwar, N. S., Snyder, P. M. & Thomas, C. P. An evolutionarily conserved N- terminal Sgk1 variant with enhanced stability and improved function. Am J Physiol Renal Physiol 295, F1440-1448, (2008). 142 Murray, J. T., Cummings, L. A., Bloomberg, G. B. & Cohen, P. Identification of different specificity requirements between SGK1 and PKBalpha. FEBS Lett 579, 991-994, (2005). 143 Inglis, S. K. et al. SGK1 activity in Na+ absorbing airway epithelial cells monitored by assaying NDRG1-Thr346/356/366 phosphorylation. Pflugers Arch 457, 1287- 1301, (2009). 144 Klingel, K. et al. Expression of cell volume-regulated kinase h-sgk in pancreatic tissue. Am J Physiol Gastrointest Liver Physiol 279, G998-G1002, (2000). 145 Warntges, S. et al. Cerebral localization and regulation of the cell volume- sensitive serum- and glucocorticoid-dependent kinase SGK1. Pflugers Arch 443, 617-624, (2002).

94

References

146 Link, W. et al. Somatodendritic expression of an immediate early gene is regulated by synaptic activity. Proc Natl Acad Sci U S A 92, 5734-5738, (1995). 147 Wulff, P. et al. Impaired renal Na(+) retention in the sgk1-knockout mouse. J Clin Invest 110, 1263-1268, (2002). 148 Huang, D. Y. et al. Blunted hypertensive effect of combined fructose and high-salt diet in gene-targeted mice lacking functional serum- and glucocorticoid-inducible kinase SGK1. Am J Physiol Regul Integr Comp Physiol 290, R935-944, (2006). 149 Farjah, M., Roxas, B. P., Geenen, D. L. & Danziger, R. S. Dietary salt regulates renal SGK1 abundance: relevance to salt sensitivity in the Dahl rat. Hypertension 41, 874-878, (2003). 150 Ullrich, S. et al. Serum- and glucocorticoid-inducible kinase 1 (SGK1) mediates glucocorticoid-induced inhibition of insulin secretion. Diabetes 54, 1090-1099, (2005). 151 Kumar, J. M., Brooks, D. P., Olson, B. A. & Laping, N. J. Sgk, a putative serine/threonine kinase, is differentially expressed in the kidney of diabetic mice and humans. J Am Soc Nephrol 10, 2488-2494, (1999). 152 Feng, Y., Wang, Q., Wang, Y., Yard, B. & Lang, F. SGK1-mediated fibronectin formation in diabetic nephropathy. Cell Physiol Biochem 16, 237-244, (2005). 153 Lang, F. et al. Deranged transcriptional regulation of cell-volume-sensitive kinase hSGK in diabetic nephropathy. Proc Natl Acad Sci U S A 97, 8157-8162, (2000). 154 Stichel, C. C. et al. sgk1, a member of an RNA cluster associated with cell death in a model of Parkinson's disease. Eur J Neurosci 21, 301-316, (2005). 155 Rangone, H. et al. The serum- and glucocorticoid-induced kinase SGK inhibits mutant -induced toxicity by phosphorylating serine 421 of huntingtin. Eur J Neurosci 19, 273-279, (2004). 156 Humbert, S. et al. The IGF-1/Akt pathway is neuroprotective in Huntington's disease and involves Huntingtin phosphorylation by Akt. Dev Cell 2, 831-837, (2002). 157 Chung, E. J. et al. Gene expression profile analysis in human hepatocellular carcinoma by cDNA microarray. Mol Cells 14, 382-387, (2002). 158 Adeyinka, A. et al. Analysis of gene expression in ductal carcinoma in situ of the breast. Clin Cancer Res 8, 3788-3795, (2002). 159 Sahoo, S., Brickley, D. R., Kocherginsky, M. & Conzen, S. D. Coordinate expression of the PI3-kinase downstream effectors serum and glucocorticoid- induced kinase (SGK-1) and Akt-1 in human breast cancer. Eur J Cancer 41, 2754-2759, (2005). 160 Shanmugam, I. et al. Serum/glucocorticoid-induced protein kinase-1 facilitates androgen receptor-dependent cell survival. Cell Death Differ 14, 2085-2094, (2007). 161 Lang, F., Perrotti, N. & Stournaras, C. Colorectal carcinoma cells--regulation of survival and growth by SGK1. Int J Biochem Cell Biol 42, 1571-1575, (2010). 162 Maiyar, A. C., Leong, M. L. & Firestone, G. L. Importin-alpha mediates the regulated nuclear targeting of serum- and glucocorticoid-inducible protein kinase (Sgk) by recognition of a nuclear localization signal in the kinase central domain. Mol Biol Cell 14, 1221-1239, (2003). 163 Kim, M. J. et al. Negative regulation of SEK1 signaling by serum- and glucocorticoid-inducible protein kinase 1. Embo J 26, 3075-3085, (2007). 164 Amato, R. et al. Sgk1 activates MDM2-dependent p53 degradation and affects cell proliferation, survival, and differentiation. J Mol Med, (2009). 165 Zhang, L., Cui, R., Cheng, X. & Du, J. Antiapoptotic effect of serum and glucocorticoid-inducible protein kinase is mediated by novel mechanism activating I{kappa}B kinase. Cancer Res 65, 457-464, (2005).

95

References

166 Amato, R. et al. IL-2 signals through Sgk1 and inhibits proliferation and apoptosis in kidney cancer cells. J Mol Med 85, 707-721, (2007). 167 Brunet, A. et al. Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor. Cell 96, 857-868, (1999). 168 Shelly, C. & Herrera, R. Activation of SGK1 by HGF, Rac1 and integrin-mediated cell adhesion in MDCK cells: PI-3K-dependent and -independent pathways. J Cell Sci 115, 1985-1993, (2002). 169 Frisch, S. M. & Francis, H. Disruption of epithelial cell-matrix interactions induces apoptosis. J Cell Biol 124, 619-626, (1994). 170 Hong, F. et al. mTOR-raptor binds and activates SGK1 to regulate p27 phosphorylation. Mol Cell 30, 701-711, (2008). 171 Dehner, M., Hadjihannas, M., Weiske, J., Huber, O. & Behrens, J. Wnt signaling inhibits Forkhead box O3a-induced transcription and apoptosis through up- regulation of serum- and glucocorticoid-inducible kinase 1. J Biol Chem 283, 19201-19210, (2008). 172 Wang, K. et al. SGK1-dependent intestinal tumor growth in APC-deficient mice. Cell Physiol Biochem 25, 271-278. 173 Nasir, O. et al. Relative resistance of SGK1 knockout mice against chemical carcinogenesis. IUBMB Life 61, 768-776, (2009). 174 Zhang, C. et al. Corticosteroids induce chemotherapy resistance in the majority of tumour cells from bone, brain, breast, cervix, melanoma and neuroblastoma. Int J Oncol 29, 1295-1301, (2006). 175 Wu, W. et al. Microarray analysis reveals glucocorticoid-regulated survival genes that are associated with inhibition of apoptosis in breast epithelial cells. Cancer Res 64, 1757-1764, (2004). 176 http://www.sigmaaldrich.com. 177 http://www.clontech.com. 178 Solinas-Toldo, S. et al. Matrix-based comparative genomic hybridization: biochips to screen for genomic imbalances. Genes Chromosomes Cancer 20, 399-407, (1997). 179 Mendrzyk, F. et al. Genomic and protein expression profiling identifies CDK6 as novel independent prognostic marker in medulloblastoma. J Clin Oncol 23, 8853- 8862, (2005). 180 Zielinski, B. et al. Detection of chromosomal imbalances in retinoblastoma by matrix-based comparative genomic hybridization. Genes Chromosomes Cancer 43, 294-301, (2005). 181 Sandoval, J. et al. Validation of a DNA methylation microarray for 450,000 CpG sites in the human genome. Epigenetics 6, 692-702, (2011). 182 Pfaffl, M. W. A new mathematical model for relative quantification in real-time RT- PCR. Nucleic Acids Res 29, e45, (2001). 183 Bukrinsky, M. I. et al. Active nuclear import of human immunodeficiency virus type 1 preintegration complexes. Proc Natl Acad Sci U S A 89, 6580-6584, (1992). 184 Blesch, A. Lentiviral and MLV based retroviral vectors for ex vivo and in vivo gene transfer. Methods 33, 164-172, (2004). 185 Moffat, J. et al. A lentiviral RNAi library for human and mouse genes applied to an arrayed viral high-content screen. Cell 124, 1283-1298, (2006). 186 Schmitt, M. & Pawlita, M. High-throughput detection and multiplex identification of cell contaminations. Nucleic Acids Res 37, e119, (2009). 187 Cory, A. H., Owen, T. C., Barltrop, J. A. & Cory, J. G. Use of an aqueous soluble tetrazolium/formazan assay for cell growth assays in culture. Cancer Commun 3, 207-212, (1991).

96

References

188 Chang, T. K., Weber, G. F., Crespi, C. L. & Waxman, D. J. Differential activation of cyclophosphamide and ifosphamide by cytochromes P-450 2B and 3A in human liver microsomes. Cancer Res 53, 5629-5637, (1993). 189 Sladek, N. E. et al. Influence of diuretics on urinary general base catalytic activity and cyclophosphamide-induced bladder toxicity. Cancer Treat Rep 66, 1889- 1900, (1982). 190 Colvin, M., Brundrett, R. B., Kan, M. N., Jardine, I. & Fenselau, C. Alkylating properties of phosphoramide mustard. Cancer Res 36, 1121-1126, (1976). 191 Friedman, O. M., Wodinsky, I. & Myles, A. Cyclophosphamide (NSC-26271)- related phosphoramide mustards- recent advances and historical perspective. Cancer Treat Rep 60, 337-346, (1976). 192 Nau, H. et al. Mutagenic, teratogenic and pharmacokinetic properties of cyclophosphamide and some of its deuterated derivatives. Mutat Res 95, 105- 118, (1982). 193 Low, J. E., Borch, R. F. & Sladek, N. E. Conversion of 4- hydroperoxycyclophosphamide and 4-hydroxycyclophosphamide to phosphoramide mustard and acrolein mediated by bifunctional catalysis. Cancer Res 42, 830-837, (1982). 194 Benckhuysen, C., van der Steen, J. & Spanjersberg, E. J. Two stable Fenton oxidation products of cyclophosphamide (NSC-26271) as precursors of 4- hydroxycyclophosphamide (NSC-196562) under physiologic conditions. Cancer Treat Rep 60, 369-372, (1976). 195 Nicoletti, I., Migliorati, G., Pagliacci, M. C., Grignani, F. & Riccardi, C. A rapid and simple method for measuring thymocyte apoptosis by propidium iodide staining and flow cytometry. J Immunol Methods 139, 271-279, (1991). 196 Freier, K. et al. Tissue microarray analysis reveals site-specific prevalence of oncogene amplifications in head and neck squamous cell carcinoma. Cancer Res 63, 1179-1182, (2003). 197 Fattet, S. et al. Beta-catenin status in paediatric medulloblastomas: correlation of immunohistochemical expression with mutational status, genetic profiles, and clinical characteristics. J Pathol 218, 86-94, (2009). 198 Herr, I. et al. Glucocorticoid cotreatment induces apoptosis resistance toward cancer therapy in carcinomas. Cancer Res 63, 3112-3120, (2003). 199 Sherk, A. B. et al. Development of a small-molecule serum- and glucocorticoid- regulated kinase-1 antagonist and its evaluation as a prostate cancer therapeutic. Cancer Res 68, 7475-7483, (2008). 200 Breitling, R., Armengaud, P., Amtmann, A. & Herzyk, P. Rank products: a simple, yet powerful, new method to detect differentially regulated genes in replicated microarray experiments. FEBS Lett 573, 83-92, (2004). 201 Foat, B. C., Morozov, A. V. & Bussemaker, H. J. Statistical mechanical modeling of genome-wide transcription factor occupancy data by MatrixREDUCE. Bioinformatics 22, e141-149, (2006). 202 Molineris, I., Grassi, E., Ala, U., Di Cunto, F. & Provero, P. Evolution of promoter affinity for transcription factors in the human lineage. Mol Biol Evol 28, 2173- 2183, (2011). 203 Remke, M. et al. FSTL5 is a marker of poor prognosis in non-WNT/non-SHH medulloblastoma. J Clin Oncol 29, 3852-3861, (2011). 204 Clifford, S. C. et al. Wnt/Wingless pathway activation and chromosome 6 loss characterize a distinct molecular sub-group of medulloblastomas associated with a favorable prognosis. Cell Cycle 5, 2666-2670, (2006).

97

References

205 Daura-Oller, E., Cabre, M., Montero, M. A., Paternain, J. L. & Romeu, A. Specific gene hypomethylation and cancer: new insights into coding region feature trends. Bioinformation 3, 340-343, (2009). 206 Mutskov, V. & Felsenfeld, G. Silencing of transgene transcription precedes methylation of promoter DNA and histone H3 lysine 9. Embo J 23, 138-149, (2004). 207 Clark, S. J. & Melki, J. DNA methylation and gene silencing in cancer: which is the guilty party? Oncogene 21, 5380-5387, (2002). 208 Stirzaker, C., Song, J. Z., Davidson, B. & Clark, S. J. Transcriptional gene silencing promotes DNA hypermethylation through a sequential change in chromatin modifications in cancer cells. Cancer Res 64, 3871-3877, (2004). 209 Zhang, X. et al. Genome-wide high-resolution mapping and functional analysis of DNA methylation in arabidopsis. Cell 126, 1189-1201, (2006). 210 Zilberman, D., Gehring, M., Tran, R. K., Ballinger, T. & Henikoff, S. Genome-wide analysis of Arabidopsis thaliana DNA methylation uncovers an interdependence between methylation and transcription. Nat Genet 39, 61-69, (2007). 211 Jones, P. A. The DNA methylation paradox. Trends Genet 15, 34-37, (1999). 212 Ball, M. P. et al. Targeted and genome-scale strategies reveal gene-body methylation signatures in human cells. Nat Biotechnol 27, 361-368, (2009). 213 Bird, A. P. & Wolffe, A. P. Methylation-induced repression--belts, braces, and chromatin. Cell 99, 451-454, (1999). 214 Downward, J. How BAD phosphorylation is good for survival. Nat Cell Biol 1, E33-35, (1999). 215 Datta, S. R. et al. Akt phosphorylation of BAD couples survival signals to the cell- intrinsic death machinery. Cell 91, 231-241, (1997). 216 Kitada, S. et al. Expression and location of pro-apoptotic Bcl-2 family protein BAD in normal human tissues and tumor cell lines. Am J Pathol 152, 51-61, (1998). 217 Chua, M. S. et al. Overexpression of NDRG1 is an indicator of poor prognosis in hepatocellular carcinoma. Mod Pathol 20, 76-83, (2007). 218 Hosoi, F. et al. N-myc downstream regulated gene 1/Cap43 suppresses tumor growth and angiogenesis of pancreatic cancer through attenuation of inhibitor of kappaB kinase beta expression. Cancer Res 69, 4983-4991, (2009). 219 Nishio, S. et al. Cap43/NDRG1/Drg-1 is a molecular target for angiogenesis and a prognostic indicator in cervical adenocarcinoma. Cancer Lett 264, 36-43, (2008). 220 Maruyama, Y. et al. Tumor growth suppression in pancreatic cancer by a putative metastasis suppressor gene Cap43/NDRG1/Drg-1 through modulation of angiogenesis. Cancer Res 66, 6233-6242, (2006). 221 Melotte, V. et al. The N-myc downstream regulated gene (NDRG) family: diverse functions, multiple applications. Faseb J 24, 4153-4166, (2010). 222 Azuma, K. et al. NDRG1/Cap43/Drg-1 may Predict Tumor Angiogenesis and Poor Outcome in Patients with Lung Cancer. J Thorac Oncol, (2012). 223 Burns, J. M. et al. A novel chemokine receptor for SDF-1 and I-TAC involved in cell survival, cell adhesion, and tumor development. J Exp Med 203, 2201-2213, (2006). 224 Murray, J. T. et al. Exploitation of KESTREL to identify NDRG family members as physiological substrates for SGK1 and GSK3. Biochem J 384, 477-488, (2004). 225 Agarwala, K. L., Kokame, K., Kato, H. & Miyata, T. Phosphorylation of RTP, an ER stress-responsive cytoplasmic protein. Biochem Biophys Res Commun 272, 641-647, (2000). 226 Desgrosellier, J. S. & Cheresh, D. A. Integrins in cancer: biological implications and therapeutic opportunities. Nat Rev Cancer 10, 9-22, (2010).

98

References

227 Kim, J. H., Zheng, L. T., Lee, W. H. & Suk, K. Pro-apoptotic role of integrin beta3 in glioma cells. J Neurochem 117, 494-503, (2011). 228 Mitra, S. K. & Schlaepfer, D. D. Integrin-regulated FAK-Src signaling in normal and cancer cells. Curr Opin Cell Biol 18, 516-523, (2006). 229 Guo, W. & Giancotti, F. G. Integrin signalling during tumour progression. Nat Rev Mol Cell Biol 5, 816-826, (2004). 230 Assoian, R. K. & Klein, E. A. Growth control by intracellular tension and extracellular stiffness. Trends Cell Biol 18, 347-352, (2008). 231 Yoo, N. J., Soung, Y. H., Lee, S. H. & Jeong, E. G. Mutational analysis of proapoptotic integrin beta 3 cytoplasmic domain in common human cancers. Tumori 93, 281-283, (2007). 232 McCubrey, J. A. et al. Roles of the Raf/MEK/ERK pathway in cell growth, malignant transformation and drug resistance. Biochim Biophys Acta 1773, 1263- 1284, (2007). 233 Melani, M., Simpson, K. J., Brugge, J. S. & Montell, D. Regulation of cell adhesion and collective cell migration by hindsight and its human homolog RREB1. Curr Biol 18, 532-537, (2008). 234 Dexamethasone, granisetron, or both for the prevention of nausea and vomiting during chemotherapy for cancer. The Italian Group for Antiemetic Research. N Engl J Med 332, 1-5, (1995). 235 Dexamethasone alone or in combination with ondansetron for the prevention of delayed nausea and vomiting induced by chemotherapy. The Italian Group for Antiemetic Research. N Engl J Med 342, 1554-1559, (2000). 236 Kirkbride, P. et al. Dexamethasone for the prophylaxis of radiation-induced emesis: a National Cancer Institute of Canada Clinical Trials Group phase III study. J Clin Oncol 18, 1960-1966, (2000). 237 Zhang, C. et al. Clinical and mechanistic aspects of glucocorticoid-induced chemotherapy resistance in the majority of solid tumors. Cancer Biol Ther 6, 278- 287, (2007).

99

Appendix

7 APPENDIX

Supplementary Table 1 Differentially expressed genes at chromosome 6q (top 50).

Gene IDs Gene Symbols Log2-ratio (6q gain/6q loss) ENSG00000118487 Q9NQ15_HUMAN 6.651536953 ENSG00000153157 SYCP2L 5.447362996 ENSG00000164483 SAMD3 5.345623716 ENSG00000112319 EYA4 4.790500393 ENSG00000135298 BAI3 4.605965507 ENSG00000188107 EGFL11 3.603930692 ENSG00000091831 ESR1 3.498036550 ENSG00000118515 SGK1 3.349049918 ENSG00000112706 IMPG1 3.059895435 ENSG00000111879 C6orf60 3.042582291 ENSG00000112305 SMAP1 2.921533071 ENSG00000135525 MAP7 2.832387863 ENSG00000135541 AHI1 2.686615866 ENSG00000025039 RRAGD 2.639821917 ENSG00000164483 SAMD3 2.613126102 ENSG00000025039 RRAGD 2.596957691 ENSG00000137343 C6orf134 2.501169734 ENSG00000111879 C6orf60 2.486942053 ENSG00000146457 WTAP 2.419393757 ENSG00000065609 SNAP91 2.372147244 ENSG00000025039 RRAGD 2.268342020 ENSG00000135314 KHDC1 2.239530546 ENSG00000096093 EFHC1 2.156483994 ENSG00000135324 C6orf117 2.105847550 ENSG00000112297 AIM1 2.091820050 ENSG00000111832 RWDD1 2.034385164 ENSG00000135541 AHI1 2.001322446 ENSG00000112305 SMAP1 1.992571868 ENSG00000118402 ELOVL4 1.961008885 ENSG00000135597 REPS1 1.948443673 ENSG00000112245 PTP4A1 1.901299836 ENSG00000196586 MYO6 1.885680113 ENSG00000153721 CNKSR3 1.884202432 ENSG00000185250 PPIL6 1.849514530 ENSG00000196911 KPNA5 1.806668531 ENSG00000132424 C6orf111 1.800259923 ENSG00000177214 C6orf224 1.779916869 ENSG00000074935 TUBE1 1.775791053 ENSG00000168216 LMBRD1 1.769003083

100

Appendix

ENSG00000112232 KHDRBS2 1.739712229 ENSG00000085382 HACE1 1.728671305 ENSG00000203756 CF191_HUMAN 1.722174483 ENSG00000119899 SLC17A5 1.719022458 ENSG00000118513 MYB 1.714005996 ENSG00000203702 CF059_HUMAN 1.709510088 ENSG00000188994 ZNF292 1.679441705 ENSG00000112238 PRDM13 1.676516373 ENSG00000079841 RIMS1 1.621929583 ENSG00000004809 SLC22A16 1.569334375 ENSG00000111880 RNGTT 1.550216488

Supplementary Table 2 Differentially expressed genes at chromosome 6q (top 50).

Gene IDs Gene Symbols ΔLog2-ratio (6q gain-6q loss) ENSG00000112232 KHDRBS2 5.44793313 ENSG00000135298 BAI3 4.79385187 ENSG00000164483 SAMD3 4.49516084 ENSG00000187772 LIN28B 4.21778024 ENSG00000118513 MYB 3.91671434 ENSG00000118495 PLAGL1 3.57651389 ENSG00000188107 EGFL11 3.48970246 ENSG00000118515 SGK1 3.42792452 ENSG00000112706 IMPG1 3.39238264 ENSG00000118487 Q9NQ15_HUMAN 3.21677199 ENSG00000213204 C6orf165 3.09191269 ENSG00000152822 GRM1 2.85350262 ENSG00000164483 SAMD3 2.83784063 ENSG00000118514 ALDH8A1 2.70925952 ENSG00000112238 PRDM13 2.69440944 ENSG00000135541 AHI1 2.68323646 ENSG00000112297 AIM1 2.64728773 ENSG00000213204 C6orf165 2.52463487 ENSG00000004809 SLC22A16 2.48251369 ENSG00000187772 LIN28B 2.36494060 ENSG00000172594 SMPDL3A 2.35684590 ENSG00000198719 DLL1 2.35627865 ENSG00000146166 GLULD1 2.35269745 ENSG00000203734 C6orf91 2.35125778 ENSG00000164442 CITED2 2.24430481 ENSG00000178425 NT5DC1 2.22621111 ENSG00000112339 HBS1L 2.22616358 ENSG00000188994 ZNF292 2.20217315 ENSG00000024862 CCDC28A 2.19786605

101

Appendix

ENSG00000118495 PLAGL1 2.13867407 ENSG00000196911 KPNA5 2.12920296 ENSG00000111912 NCOA7 2.11746298 ENSG00000049618 ARID1B 2.09846149 ENSG00000085382 HACE1 2.09128243 ENSG00000083097 DOPEY1 2.09080476 ENSG00000177214 NP_775830.1 2.08627999 ENSG00000116183; PAPPA2;PTP4A1 2.06406025 ENSG00000112245 ENSG00000026652 AGPAT4 2.04941124 ENSG00000135597 REPS1 2.04364622 ENSG00000065609 SNAP91 2.01824604 ENSG00000112486 CCR6 2.01054462 ENSG00000146263 C6orf167 2.00165799 ENSG00000196586 MYO6 1.99995427 ENSG00000154548 SRR35_HUMAN 1.94918374 ENSG00000168438 CDC40 1.94375934 ENSG00000196586 MYO6 1.91612585 ENSG00000146411 SLC2A12 1.90527704 ENSG00000111912 NCOA7 1.88184449 ENSG00000118496 FBXO30 1.87804212 ENSG00000135314 C6orf148 1.85990309

Supplementary Table 3 Top 50 (total 172) upregulated genes upon SGK1 knockdown, fold change (knockdown/control) value indicates the mean over all experiments for respective probe.

Agilent IDs Gene Symbols Fold-change (Log2-ratio) A_24_P337239 FGF5 2,6875 A_24_P158089 SERPINE1 2,4275 A_23_P101054 KRT34 2,1970 A_23_P333228 MARCH4 2,0584 A_23_P72096 IL1A 2,0097 A_24_P119201 MBD2 1,9364 A_23_P69810 AGPAT9 1,9162 A_23_P123234 SERPINE1 1,9051 A_24_P143118 SCML2 1,8548 A_23_P79518 IL1B 1,7668 A_23_P156826 C6orf105 1,7613 A_23_P215956 MYC 1,7131 A_23_P213319 ADAMTS6 1,6842 A_23_P136493 NRG1 1,6787 A_24_P397204 KCNIP4 1,6687 A_23_P76078 IL23A 1,6616 A_24_P926284 OFCC1 1,6374 A_23_P317760 C6orf146 1,6128 A_23_P212800 FGF5 1,6110

102

Appendix

A_32_P87013 IL8 1,5719 A_23_P106389 SEMA7A 1,5225 A_23_P214821 EDN1 1,5102 A_24_P355057 SLC13A1 1,5035 A_24_P936899 C12orf63 1,5020 A_23_P159325 ANGPTL4 1,5002 A_23_P38519 ITGB3 1,4639 A_24_P410498 WDFY4 1,4600 A_24_P198044 ZNF483 1,4452 A_24_P915734 C2orf82 1,4335 A_23_P111311 AKAP12 1,4285 A_23_P135381 SP5 1,4239 A_32_P88555 ATF7 1,4215 A_23_P118842 KRTAP1-5 1,4111 A_24_P33895 ATF3 1,4065 A_23_P34915 ATF3 1,3969 A_23_P338479 CD274 1,3958 A_23_P315815 NRG1 1,3764 A_24_P193011 CCND1 1,3752 A_23_P403488 NLRP10 1,3587 A_24_P247738 DNAH8 1,3440 A_23_P156861 RGS17 1,3389 A_23_P214897 AKAP12 1,3278 A_23_P25194 HRK 1,3267 A_24_P459739 1,3262 A_24_P307885 ERGIC1 1,3092 A_32_P196193 PAQR9 1,3032 A_24_P255233 AC100793.1 1,2998 A_24_P14902 TAS2R42 1,2962 A_23_P34366 HTR1D 1,2961 A_23_P43197 CALB1 1,2937

Supplementary Table 4 Top 50 (total 288) downregulated genes upon SGK1 knockdown, fold change (knockdown/control) value indicates the mean over all experiments for respective probe.

Agilent IDs Gene Symbols Fold-change (Log2-ratio) A_23_P204286 MGP -3,3865 A_23_P86470 CH25H -3,0962 A_23_P170649 C8orf84 -3,0471 A_23_P154605 SULF2 -3,0376 A_24_P173823 PBX1 -3,0123 A_23_P257307 SERPINA7 -2,9252 A_23_P113351 SPARCL1 -2,8888 A_23_P109322 PCP4 -2,8818 A_23_P83134 GAS1 -2,8370

103

Appendix

A_23_P91230 SLPI -2,8163 A_24_P190472 SLPI -2,7682 A_32_P134764 CDH6 -2,7134 A_32_P188860 IL17RD -2,6920 A_23_P206022 ITGA11 -2,6799 A_23_P43164 SULF1 -2,6773 A_23_P383009 IGFBP5 -2,6309 A_23_P58676 C5orf23 -2,6214 A_23_P110403 PDLIM3 -2,6205 A_23_P213424 ENC1 -2,5899 A_23_P6263 MX2 -2,5460 A_23_P75299 LHPP -2,5200 A_23_P201459 IFI6 -2,5186 A_23_P19673 SGK1 -2,4970 A_23_P34126 BGN -2,4882 A_23_P24507 AP002004.1 -2,4737 A_24_P277934 COL1A2 -2,4703 A_23_P17345 MAFB -2,4673 A_24_P347411 POSTN -2,4590 A_23_P211631 FBLN1 -2,4555 A_23_P28307 DHX57 -2,4527 A_24_P306814 ASS1P9 -2,4526 A_23_P61508 CHRNA9 -2,4487 A_23_P2492 C1S -2,4165 A_23_P137016 SAT1 -2,4161 A_23_P121533 SPON2 -2,3892 A_23_P216361 COL14A1 -2,3887 A_23_P46045 RGS5 -2,3827 A_24_P130363 C18orf1 -2,3659 A_23_P18447 PPARGC1A -2,3626 A_24_P246825 C6orf138 -2,3340 A_32_P155026 NFIA -2,3294 A_23_P207003 SEPT_4 -2,3292 A_23_P371495 TMTC1 -2,3257 A_23_P3911 PLXDC1 -2,3037 A_23_P127584 NNMT -2,2910 A_23_P150162 DRD4 -2,2905 A_23_P337262 APCDD1 -2,2844 A_23_P90189 BBC3 -2,2805 A_23_P143247 TSHZ2 -2,2713 A_32_P108254 FAM20A -2,2708

104

Acknowledgements

8 ACKNOWLEDGEMENTS

My sincere gratitude goes to Prof. Peter Lichter and Prof. Stefan Pfister for giving me the opportunity to conduct my PhD thesis in such a fruitful and inspiring surrounding, for the supervision and lively discussions, great ideas, general support, and proof reading of my thesis.

Furthermore, I would like to thank Prof. Werner Buselmaier for heading my examination, for all his support during my time as a PhD student and for being a member of my thesis advisory and examination committee.

I would especially like to thank Dr. Marcel Kool for great input in discussions, continuous support and advice, and for the proofreading of my thesis introduction.

Moreover, I very much appreciate the willingness of Prof. Stephan Frings and Prof. Peter Angel to be part of my examination committee.

I thank Prof. Olaf Witt and Prof. Michael Boutros for their support as members of my thesis advisory committee (TAC).

Particularly, I want to thank Dominik Sturm for co-working on this project, focusing on the clinical aspects and his never ending support and great discussions on lab issues as well as beyond the lab.

I would like to thank our collaboration partners Prof. Florian Lang and Dr. Eva-Maria Schmidt from the university of Tübingen for helping with the migration experiments and providing an SGK1 antibody, and Dr. Apfel for transporting the cells from Heidelberg to Tübingen.

Moreover, I would like to thank my collaborators, in particular Prof. Andrey Korshunov, Marina Ryzhova for their neuropathologic expertise, Dr. Marc Remke, Dr. Hendrik Witt, and Dr. David Jones for excellent collaboration.

105

Acknowledgements

Dr. Marc Zapatka and Dr. Rosario Piro I would like to thank for conducting all statistical analyses I was not able to do on my own.

For excellent technical assistance, especially in the cell culture, I am also grateful to Linda Linke. Moreover I would like to thank Andrea Wittmann, Magdalena Schlotter, Laura Sieber, and Laura Puccio for always having a helping hand whenever it was needed.

Beyond that, I thank Sonja Hutter, Theo Tzaridis, Huriye Cin, Elke Pfaff, and Michael Hain for universal support in labwork/IT problems and being great lab mates.

For help in experimental issues and fruiful discussions in and outside the lab I would like to thank Dr. Thorsten Kolb, Dr. Jan Gronych, Dr. Daniel Haag, Dr. Martje Tönjes, and Laura Dittmann.

For the nice atmosphere and being valuable companions of my time during and beyond the lab I would like to thank Sebastian Bender, Dr. Angela Schulz, Verena Thewes, Josy Bageritz, Katharina Filarsky, Dr. Agata Olszak, and Claudia Dürr.

For being wonderful friends and supporting me in every circumstance, I deeply thank Dr. Inn Chung, Elle Wieland, Dr. Alexandra Georgi, Anne Ferracane, and Dr. Leah Wallenta.

Finally and most importantly, I deeply thank my parents Monika and Gerhard, my sister Milena and my grandma Hedwig for their unconditional support, appreciation and constant believe in me. You provided me with confidence and motivation helpful for this time.

106