" Deletion and Functional Analysis of Fetuin-B"

Von der Fakultät für Mathematik, Informatik und Naturwissenschaften der Rheinisch-Westfälischen Technischen Hochschule Aachen zur Erlangung des akademischen Grades einer Doktorin der Naturwissenschaften genehmigte Dissertation

vorgelegt von

Diplom-Biologin

Jennifer Wessling geb. Goldstein

aus Aachen

Berichter: Universitätsprofessor Dr. rer. nat. W. Jahnen-Dechent Universitätsprofessor Dr. rer. nat. L. Elling

Tag der mündlichen Prüfung: 21. November 2007

Diese Dissertation ist auf den Internetseiten der Hochschulbibliothek online verfügbar.

Contents

1 Introduction 5 1.1 Fetuin Family of ...... 5 1.1.1 Structure ...... 6 1.1.2 Expression ...... 10 1.1.3 Genetic Organization ...... 11 1.1.4 Biological Role ...... 12 1.2 Female Reproduction ...... 14 1.2.1 Oogenesis and Ovulation ...... 14 1.2.2 Fertilization ...... 16 1.2.3 Parthenogenesis ...... 17 1.3 The Mouse as a Model System ...... 17 1.3.1 Targeting the Mouse Genome ...... 18 1.4 Aim of the Study ...... 20

2 Materials and Methods 21 2.1 Oligonucleotides ...... 22 2.2 Generating a fetuin-B-deficient Mouse Strain ...... 23 2.2.1 Cloning of the Gene Targeting Vector ...... 23 2.2.2 Gene Targeting in Embryonic Stem Cells ...... 25 2.2.3 DNA Extraction from Cells in 96-Well Plates . . . . . 28 2.2.4 PCR Screening ...... 28 2.2.5 Southern Blot ...... 30 2.2.6 Blastocyst Injection ...... 30 2.2.7 Embryo Transfer ...... 30 2.2.8 Animal Husbandry and Breeding ...... 31 2.2.9 Genotyping ...... 31 2.3 Analysis of fetuin-B-deficient Mice ...... 32

i 2.3.1 Western Blot ...... 32 2.3.2 Immunohistochemistry ...... 33 2.3.3 Collecting Mouse ...... 34 2.3.4 Primary Morphological Analysis ...... 35 2.3.5 Necropsy ...... 35 2.3.6 Histology ...... 36 2.3.7 Weighing ...... 37 2.3.8 Blood Pressure Measurement ...... 37 2.3.9 Glucose Tolerance Test ...... 37 2.3.10 Clinical Chemical Screening ...... 38 2.4 Recombinant Expression and Purification ...... 38 2.4.1 Cloning Murine Fetuin-B ...... 38 2.4.2 Generating a Fetuin-B Transfecting Adenovirus . . . . 41 2.4.3 Expression and Purification of Fetuin-B ...... 41 2.5 In Vitro Analysis of Fetuin-B ...... 42 2.5.1 Precipitation Inhibition Assay ...... 42 2.5.2 Mink Lung Cell Reporter Assay ...... 43 2.6 Promoter Analysis ...... 44 2.6.1 Cloning of Reporter Constructs ...... 44 2.6.2 Transfection of Reporter Constructs ...... 44

3 Results 47 3.1 Generating a Fetuin-B-deficient Mouse Strain ...... 47 3.1.1 Gene Deletion and the Targeting Vector ...... 47 3.1.2 Establishing the Screening Methods ...... 50 3.1.3 Gene Targeting in Embryonic Stem Cells ...... 52 3.1.4 Screening for Recombinant ES Cell Clones ...... 54 3.1.5 Blastocyst Injection and Chimera ...... 55 3.1.6 Embryo-Transfer and Breeding ...... 57 3.1.7 Confirmation of Fetuin-B-deficient Mice ...... 58 3.2 Phenotyping Fetuin-B-deficient Mice ...... 59 3.2.1 Fertility ...... 60 3.2.2 Anatomical and Morphological Analysis ...... 67 3.2.3 Physiological Parameters ...... 74 3.2.4 Glucose Tolerance Test ...... 74 3.2.5 Clinical Chemical Screening ...... 78

ii 3.2.6 Summarizing the Phenotyping Results ...... 80 3.3 Recombinant Murine Fetuin-B ...... 81 3.3.1 Cloning Recombinant Murine Fetuin-B ...... 81 3.3.2 Generating a Fetuin-B Transfecting Adenovirus . . . . 86 3.3.3 Expressing and Purifying Recombinant Fetuin-B . . . . 86 3.3.4 Precipitation Inhibition Assay ...... 90 3.3.5 TGF-β Reporter Activity in Mink Lung Cells . . . . . 90 3.3.6 Serum Concentration of Fetuin-B ...... 93 3.3.7 Summarizing the Results of the in Vitro Data . . . . . 94 3.4 Promoter Analysis ...... 95 3.4.1 Cloning and Analysis of the Fetub Promoter ...... 95

4 Discussion 99 4.1 Generation of Fetuin-B-deficient Mice ...... 99 4.1.1 Gene Deletion ...... 99 4.1.2 Embryonic Stem Cells and Genetic Background . . . . 99 4.1.3 Screening Procedure ...... 100 4.1.4 Expression of Fetuin Family Proteins ...... 101 4.1.5 Fetuin Serum Concentrations ...... 101 4.2 Phenotyping ...... 102 4.2.1 Vitality ...... 102 4.2.2 Female Infertility ...... 102 4.2.3 Anatomy and Morphology ...... 106 4.2.4 Physiology and Learning ...... 107 4.2.5 Clinical Chemistry Screening ...... 108 4.3 Recombinant Murine Fetuin-B ...... 109 4.3.1 Eukaryotic Expression of Recombinant Fetuin-B . . . . 109 4.3.2 Fetuin-B Transfecting Adenovirus ...... 110 4.3.3 Inhibition of Ectopic Calcification ...... 110 4.3.4 TGF-β Biology ...... 111 4.4 Outlook ...... 112

Summary 113

Appendix 115 Pedigrees ...... 115 Fetub Genomic Sequence ...... 117

iii Abbreviations ...... 125 List of Figures ...... 128 List of Tables ...... 129

References 142

Acknowledgements 143

Curriculum Vitae 145

iv Chapter 1

Introduction

1.1 Fetuin Family of Proteins

For a long time the fetuin family of proteins consisted of a set of orthologous serum proteins in humans, sheep, pigs, cows and rodents but also in birds and reptiles. They were variably designated fetuin in sheep, pig and cow, α2-HS- (AHSG) in human, phosphoprotein of 63 kDa (pp63) in rat or countertrypin in mouse and gerbil. However, their close sequence ho- mology, especially a set of 12 cysteine residues at fixed distances and a fetuin motif (LETXCHXLDPTP), indicated that fetuin, AHSG, pp63 and coun- tertrypin emerged from a single ancestral gene and resembled counterparts (orthologous) among these species [1, 2, 3, 4, 5]. The fetuins were classified as cystatin superfamily members because they contain cystatin-like domains characterized by the distinct cysteine residues. Further members of the cystatin superfamily, all sharing cystatin-like do- mains, are the cystatins themselves, histidine-rich (HRG) and kininogens (KNG). The cystatins are low-molecular-weight cysteine protease inhibitors and prototypic for the cystatin superfamily [6, 7]. A study that aimed at systematic cloning of hepatic mRNAs corresponding to inflammation-related in rat [8] revealed a second member of the fetuin family based on sequence similarity. Sequence analysis showed that the novel protein contained both fetuin signatures, including the set of 12 cysteine residues at fixed distances, perfectly conserved, and the fetuin motif that is present in a truncated version (LETGCHVL) [9]. The novel fetuin, fetuin- B was found in rat, mouse and humans, revealing both fetuin signatures

5 6 CHAPTER 1. INTRODUCTION with a 61% overall (strictly conserved residues) in these species. When compared to the original fetuin, fetuin-A, fetuin-B shared 17% of strictly conserved amino acids and 24% of similar residues. The similarity was significant but much lower than the similarity between the rat, mouse and human fetuin-A (58% strictly conserved residues). The sequence analysis indicated that fetuin-B was homologous, yet, clearly different from fetuin-A. Therefore, the novel fetuins were considered a novel entity within the fetuin family. The two paralogous were designated fetuin-A (comprising fetuin, AHSG, pp63 and countertrypin) and fetuin-B (alias fetuin beta) [9].

1.1.1 Protein Structure

Three major domains (D1-D3) characterize fetuin proteins. Domain D1 and D2 are tandemly arranged cystatin-like domains containing 12 cysteine residues at fixed distances, and are followed by a unique domain, D3, that is only loosely conserved between species [10]. In fetuin-A, domains D1 and D2 both contain dibasic motifs, db1 and db2 respectively. They are thought to account for protease-inhibition domains at least in human and bovine fetuin-A. Additionally, domain D1 contains a calcium, TGF-β and lectin binding sites [1, 11, 12]. Domain D3 can further be subdivided into D3a, a hydrophobic, proline-rich N-terminal half, and D3b, a C-terminal half including a connecting peptide, at least in humans [1, 7, 13, 14]. The general organization of domains is maintained in fetuin-B, as shown in Figure 1.1. However, the dibasic motifs db1 and db2 are only loosely retained and the calcium-binding site in domain D1 is absent. Furthermore, domain D3 does contain a proline-rich N-terminal but no connecting peptide in the C-terminal half. However, fetuin-B contains a short homology to the TGF-β receptor type II (depicted in Figure 1.2) and two archetypal Kunitz- type motifs, associated with protease-inhibitory activity, within domain D1 and D2 that is only present in fetuin-A domain D2 [9]. However, none of these domains were functionally verified. 1.1. FETUIN FAMILY OF PROTEINS 7 8 CHAPTER 1. INTRODUCTION

Figure 1.1: Alignment of rat, mouse and human fetuin-B (f-B) with fetuin- A (f-A) from various mammals (Olivier et al. [9]). White residues on black background indicate identical or similar residues conserved in at least 10 out of 11 sequences. Hyphens indicate gaps; boxes represent the proposed signal peptides. The LETXCHXLDPTP fetuin signature is underlined. The 12 crit- ical cysteine residues are marked with a dot. A CPG tripeptide conserved in the cystatin superfamily is double-underlined. Disulphide bonds are indicated by a horizontal connecting line between cysteine residues; the bond connecting the first and ultimate cysteine is noted with a dashed line: The domains D1, D2 and D3 (D3a and D3b) are marked with broken arrows. The connecting peptide is over-lined and marked with CP. The potential calcium-binding site is over-lined and marked with CBS. The dibasic peptides db1 and db2 are over-lined. 1.1. FETUIN FAMILY OF PROTEINS 9

Figure 1.2: Modeled binding of human TGF-beta to its receptor type II (structure software Pymol 0.9V). The TGF-β binding site is depicted in green. The homology between the receptor and fetuin-B is depicted in yellow. Note that it is not part of the TGF-β binding site. 10 CHAPTER 1. INTRODUCTION

1.1.2 Expression

On the mRNA Level

The tissue-specific expression pattern of fetuin-A and -B mRNA is similar. In all cases, fetuin mRNA is predominantly expressed by the liver. However, fetuin-A mRNA can be detected to a much lower extent in the kidney, embryo and tongue, whereas fetuin-B mRNA expression is additionally expanded to the lung and ovary [15]. Fetuins are differentially expressed during development. In rodents the fetuin-A liver mRNA level exhibits a perinatal and transient increase during early post-natal life while this only happens for fetuin-B in the mouse. In the rat, the mRNA expression of fetuin-B plateaus and is retained in adulthood. Furthermore, fetuins have been shown to be subject to inflammation- associated changes in hepatic mRNA expression. Fetuin-A and -B are neg- ative acute-phase proteins, meaning that their hepatic expression is down- regulated in response to an inflammatory stimulus [16, 17].

On the Protein Level

Fetuins are abundant serum proteins. Unlike fetuin-A, fetuin-B expression is twice as high in human females as in males. Like most serum proteins, fetuins penetrate most soft tissues. In this respect, the tissue distribution of fetuin- A and -B is similar, but the amounts vary within the given tissue. Fetuin-A is present in the cecum, heart, placenta, skin and tongue in higher amounts than in the colon, liver, lung, skeletal muscle, small intestine, spleen and stomach. In comparison, fetuin-B is present in the cecum, kidney, placenta, skin and tongue in higher amounts than in the colon, heart, liver, lung, skeletal muscle, small intestine, spleen and stomach [15]. Two distinct forms of fetuin-B were detectable at least in the liver and tongue. There was evidence that these reflected secondary modifications, including limited proteolysis, differential glycosylation and phosphorylation within the highly-expressive fetuin-B mRNA tissues [15]. However, others propose the existence of fetuin-B splice variants [18]. 1.1. FETUIN FAMILY OF PROTEINS 11

1.1.3 Genetic Organization The genes for fetuin-A (Ahsg) and fetuin-B (Fetub) are single-copy genes and direct neighbors on mouse 16 and human chromosome 3, followed by other members of the cystatin superfamily Hrg and Kng as shown in Figure 1.3.

[22857939 > [23029090 >

Dnajb11 Ahsg Fetub Hrg Kng2

Figure 1.3: Genomic context of fetuin-A (Ahsg) and fetuin-B (Fetub) on mouse (from NCBI, Gene).

Figure 1.4 illustrates the genetic organization of the fetuin genes in the mouse. The gene for fetuin-A consists of seven exons and six introns. The first exon begins with an untranslated region (UTR) followed by the cod- ing region. The coding sequence ends in the last exon and is followed by a 3’UTR. Exons 1 to 3 code for the two cystatin-like domains D1 and D2.

Figure 1.4: Transcripts and products of Ahsg and fetuB genes in the mouse. Modified figure from NCBI, Entrez Gene. Exons are numbered. Reference- nucleotide sequences are written in blue letters, protein sequences in red let- ters. 12 CHAPTER 1. INTRODUCTION

The genetic organization of fetuin-B is different from that of fetuin-A. The fetuin-B gene consists of eight exons. The first exon contains the 5’UTR only. The coding sequence does not start until exon 2 [9]. However, two alternative first exons encoding distinct 5’UTRs exist. These alternative transcripts were designated isoforms 1 and 3. Isoform 2 uses the same 5’UTR as isoform 1, but isoform 2 lacks exon 2 due to alternative splicing. However, this data was derived from in silico cloning and needs experi- mental confirmation.

1.1.4 Biological Role

Fetuin-A was first described in 1944 by Pedersen [19] as the most abundant globulin in fetal bovine serum. Since then many distinct biological functions have been proposed for fetuin-A, indicating its interaction with many diverse target molecules. Like serum albumin, α2-macroglobulin and apolipoproteins that are well-established carrier proteins, the binding and sequestration of otherwise insoluble ligands also seem to be major functions of fetuin-A. In brief, fetuin-A has been shown to influence opsonization, lipid trans- port, cell proliferation, inhibition of the insulin receptor, protease inhibition and hematopoietic cell homing [2]. Additionally, fetuin-A has been shown to interfere with transforming growth factor-β (TGF-β), bone morphogenetic protein (BMP) and hepatocyte growth factor (HGF) binding to their signal- ing receptors [12, 20]. Furthermore, animal studies have revealed a possible interference of fetuin-A with the signaling of lipopolysaccharide (LPS) and tumor necrosis factor-α (TNF-α) [21, 22, 23]. Fetuin-A contains a mineral-binding structure that binds apatite micro- crystals. Thus, fetuin-A inhibits calcification in the extracellular fluid, which is a metastable system with regard to calcium and phosphate ions [11, 24]. The physiological function of fetuin-B needs to be elucidated. The struc- tural homology and expression patterns of fetuin-A and -B indicate similar but also distinct functions [15]. The structural analysis of fetuin-B revealed no functional calcium-binding domain [11]. Genetic association studies sug- gest a role for fetuin-B in tumor biology [18]. However, the underlying molec- ular function has not yet been revealed. In summary, a clear physiological function for fetuin-B was lacking at the start of this work. 1.1. FETUIN FAMILY OF PROTEINS 13

Fetuin-A-Deficient Mouse Model

Jahnen-Dechent et al. generated a mutant mouse strain deficient in fetuin- A. These animals were vital, and fertile [25]. The calcification-inhibitory function of fetuin-A was proven in this mouse strain by Sch¨afer et al. [24]. The fetuin-A-deficient mice revealed a mild calcification phenotype [25] or a severe systemic calcification of vital organs, dependent on the genetic back- ground, C57BL/6 and DBA/2 respectively. The influence of the genetic background suggests that additional serum proteins may compensate for the loss of fetuin. However, a hypercalcemic diet induced a soft-tissue calcifi- cation phenotype in calcification resistant mouse strains, revealing the im- portance of fetuin-A within the pool of inhibitory proteins. The phenotype resembled calciphylaxis, a particularly severe calcifying disorder in end-stage renal disease patients. In this respect, these animals were employed to study the impact of fetuin-A on vascular calcification and atherosclerosis in chronic kidney disease in vivo [26, 27]. Besides the defective inhibition of apatite formation in serum, a study on metabolism in fetuin-A-deficient mice revealed that fetuin-A deficiency contributed to the decreased body weight associated with aging and under normal and high-fat diets. Furthermore, the level of circulating free fatty acids, triglycerides and insulin was shown to be reduced. This was accom- panied by improved glucose tolerance in the fetuin-A-deficient mice [28, 29]. Induced tumor studies on the fetuin-A-deficient mouse model implied fetuin-A’s role in tumor genesis and progression. Fetuin-A contributed to early stages of skin tumorigenesis [30], to the progression of TGF-β-dependent tumors [31], and to the promotion of metastasis of injected Lewis lung carci- noma (LLC) cells in vivo [32], indicating that fetuin-A contributes to tumor promotion and suppression depending on tumor stage and entity. However, neither C57BL/6 nor DBA/2 fetuin-A-deficient mice were particularly prone to spontaneous tumor formation. The calcification-inhibitory potential of fetuin-A was proven by the most prominent phenotype of the fetuin-A-deficient mice: ectopic calcification. Moreover, the fetuin-A-deficient mice offer an excellent opportunity to prove the in vivo relevance of in vitro generated data on the biological function of fetuin-A. Up to now, there was no fetuin-B-deficient mouse model available to study 14 CHAPTER 1. INTRODUCTION the physiological function of fetuin-B in a biological system. This work de- scribes the first fetuin-B-null mutant mouse model. The generation of a fetuin-B-deficient animal model and its characterization will be described in the following chapters. It will be shown that fetuin-B-deficient females were infertile.

1.2 Female Reproduction

1.2.1 Oogenesis and Ovulation

In mice, primordial germ cells enter the gonads at 11.0 days post coitus (dpc). Follicle formation begins perinatally when germ cell cysts are invaded by pre-granulosa cells to establish individual primordial follicles. Each is contained within one follicle surrounded by the granulosa cells that contribute to oocyte growth and differentiation [33]. By 5 days after birth, all oocytes are diploid and contain four times the haploid amount of DNA. They are arrested in the diplotene stage of the prophase of the first meiotic division [34]. During this time the maternal mRNA is transcribed that is required for oogenesis and embryonic develop- ment [35, 36]. When females reach sexual maturity, during each reproductive cycle, primordial follicles are recruited from the resting pool (104 primordial follicles) to start a growth phase [37]. The growing oocyte, preceding ovulation, deposits a layer of extracellular matrix, the (ZP) around the oocyte [38, 39]. It contributes to oocyte growth, serves as a polyspermy block and promotes pre-implantation development [40]. The murine ZP is composed of three sulfated glycopro- teins: ZP1, ZP2, and ZP3 [41]. ZP2 and ZP3 resemble long filaments inter- connected to each other by ZP1 [42, 41]. ZP3 binds free-swimming sperm during fertilization [43] and triggers the acrosome reaction that is essential for ZP penetration and subsequent fertilization. ZP2 is modified after fertiliza- tion to prevent polyspermy. This process is called zona pellucida hardening [44]. A hormonal stimulus provided by the pituitary, the increase of follicle stimulating hormone (FSH), causes the oocyte to resume the final stages of meiosis I. Only a few follicles respond to the release of FSH in each repro- ductive cycle, which occurs every four days. The responding follicles develop 1.2. FEMALE REPRODUCTION 15 into antral (graafian) follicles, in which the stimulated follicle cells break con- tact with the oocyte and increase their production of high-molecular-weight proteogycans and tissue plasminogen activator. Meanwhile, the follicle ac- cumulates fluid, swells, and moves toward the periphery of the ovary [44]. Just before ovulation, a surge in luteinizing hormone (LH) causes nuclear (germinal vesicle) maturation of the oocyte, preceding nuclear breakdown (germinal vesicle breakdown) and the first meiotic division. This process is completed by the extrusion of the first polar body containing one set of the homologous and a small amount of cytoplasm. The other set of chromosomes remains in metaphase II and only resumes meiosis after fertilization. Oocytes arrested in metaphase II are ovulated surrounded by a mass of follicle cells. This aggregate, associated by the previously secreted proteo- glycans, is called the cumulus oocyte complex (COC). The follicle cells that remain in the ovary differentiate into steroid-secreting cells that contribute to maintaining the pregnancy.

Figure 1.5: Micrograph of an ovulated mouse oocyte at day 0.5 pc. 40x magnification, light microscopy with differential interference optics. 16 CHAPTER 1. INTRODUCTION

8-12 oocytes are usually ovulated during one reproductive cycle. This process is asynchronous and requires 2-3 hours. The COC is transported by epithelial cilia into the open end of the oviduct called infundibulum. The section of the tube adjacent to the infundibulum enlarges and forms the ampulla where fertilization takes place.

1.2.2 Fertilization

Once the COC reaches the ampulla, it is exposed to the ejaculated sperm. Capacitated sperm first need to penetrate through the cumulus mass before they reach the zona pellucida. As described earlier, sperm bind to ZP3 within the ZP, most probably to specific O-linked oligosaccarides attached to the ZP3 polypeptide [45, 46]. ZP3-binding triggers the acrosome reaction. This process releases hydrolytic enzymes from a secretory-vacuole-like structure, the acrosome, that is located in the sperm’s head. The sperm’s acrosome reaction is essential for penetrating the ZP and for subsequent fusion with the oocyte plasma membrane [40]. Sperm that penetrate through the ZP bind to oocyte-cell-surface molecules in order to fuse with the oocyte plasma membrane. Members of the ADAM family on sperm and integrins on the oocyte were thought to be partners for the binding and fusion of the plasma membranes [47, 48, 49]. More recently, CD9, a member of the tetraspanin superfamily of integral plasma membrane proteins that associates with integrins on the oocyte surface, was shown to play a role in sperm-oocyte fusion [50, 51, 52], and PSG17, a member of the immunoglobulin superfamily, may serve as a binding partner on sperm [53]. The fusion of sperm and oocyte triggers a cascade of reactions known as fertilization. Polyspermy is excluded by the immediate change in the oocyte surface to inhibit further fusions. Furthermore, the Ca++-dependent release of cortical granules initiates the zona pellucida reaction in which the ZP glycoproteins are further cross-linked, leading to zona pellucida hardening, another block to polyspermy. At the same time, ZP3 looses its ability to bind sperm [44]. Moreover, fertilization triggers the resumption of meiosis. The maternal sister chromatids that were arrested in the metaphase II stage are now sep- arated and extruded as the second polar body. Two distinct pronuclei are formed containing the haploid maternal and paternal chromosomes. While 1.3. THE MOUSE AS A MODEL SYSTEM 17 both sets of haploid chromosomes are being replicated, the pronuclei move toward the center of the oocyte. This involves modifications of the cytoskele- ton. The membranes break down (germinal vesicle breakdown) and the chro- mosomes assemble on the spindle. Soon thereafter, the first cleavage occurs: the oocyte-to-embryo transition [44].

1.2.3 Parthenogenesis

Murine oocytes can initiate embryonic development in the absence of sperm. This process is called parthenogenesis. The mouse strain LT/Sv is prone to parthenogenetic activation (about 10% of oocytes). These embryos develop to the gastrula stage at 7 dpc. before they die. Parthenogenetic activation can also be induced, e.g. by deleting the c-mos gene [54, 55] or by exposing the oocytes to activating agents such as alcohol, hyaluronidase, Ca++/Mg++- free medium, heat or cold shock, and anesthetics.

1.3 The Mouse as a Model System

The challenge to understanding the heredity of mammalian organisms was the driving force that led to the science of genetics. Knowledge about how genes control mammalian development was first attained by studying the inheritance of coat color in a variety of domestic animals. Of all mammals studied by the early geneticists at the beginning of the 20th century, the house mouse (Mus musculus) became the animal of choice because of its size, resistance to infection, large litter size, rapid generation time and the pool of mutations affecting coat color and behavior that were available. Today large amounts of genetic information on the mouse have accumu- lated. The mouse genome has been sequenced and assembled. A physical map of the entire murine genome is available on nearly 300 bacterial artificial chromosomes. It has been shown that about 99% of the murine genes have a human homolog. This has made mus musculus a model for human genetics and disease. Table 1.1 provides some statistical information about this model organism. The ability to manipulate the genome of the mouse, which was established in the 1980s, has once more contributed to the experimental significance of the mouse. The targeted mutation of specific genes by means of homologous 18 CHAPTER 1. INTRODUCTION

Table 1.1: Vital statistics of the European House Mouse (Mus musculus).

Genome Number of chromosomes 40 Diploid DNA content 2.6·109bp Approximate number of genes 30,000-46,000 Highly repeated DNA sequences 8-10%

Reproductive biology Gestation time 19-20 days Age at weaning 21 days Age at sexual maturity 6-8 weeks Approximate weight birth 1 g weaning 8-12 g adult 30-40 g (female

1.3.1 Targeting the Mouse Genome

In classical genetics unspecifically induced mutations were used for the func- tional analysis of genes in vivo. The generation of knockout mice by means of gene targeting, meaning the site-directed intermolecular homologue recom- bination of DNA, was introduced in 1988 by Capecchi [56]. A series of events had previously facilitated this technique such as the isolation and culture of murine embryonic stem cells derived from the inner cell mass of blastocysts in 1981 [57, 58] and the successful homologous re- 1.3. THE MOUSE AS A MODEL SYSTEM 19 combination of foreign DNA into the genome of mammalian fibroblasts in 1985 [59]. The injection of ES cells into the cavities of blastocysts introduces the genetic alterations which have been performed in the ES cells into the mouse embryo. The manipulated ES cells develop together with the cells of the inner cell mass of the blastocyst to form a chimeric pup that is delivered by a foster mother. The contribution of the manipulated ES cells is revealed by the chimera’s coat color when the manipulated ES cells and the recipient blastocyst are derived from different mouse strains. E.g. ES cells contain the dominant agouti gene while blastocysts carry the recessive black gene. After the mating of the chimera with a black mouse, germ-line-transmitting chimeras give rise to agouti and black colored mice in the F1 generations, an apparent contradiction to Mendel’s law of uniformity. The agouti mice descend directly from the injected ES cells. Statistically 50% of them are heterozygous for the targeted mutation. Mice homozygous for the targeted mutation are obtained by mating het- erozygous F1 litter mates. The F2 generation typically consists of 50% het- erozygous, 25% wildtype and 25% mice homozygous for the targeted muta- tion according to Mendel’s law of segregation. If hybrid stem cells were used, a series of backcrossing events with an inbred strain needs to be performed to obtain a uniform genetic background. Inbred strains are identical at all genetic loci and thereby provide insight into the impact of the gene deletion on the phenotype.

The Gene Targeting Vector

The aim of gene targeting is the loss of an essential DNA sequence leading to a non-functional gene. This technique is based on homologous recombina- tion. The targeting vector contains the homologue DNA sequence, a positive selection marker that replaces the essential DNA sequence within the ho- mologous sequence and a negative selection marker flanking the homologous sequence. After transfection, the gene-targeting vector binds to the genomic ho- mologue sequence and replaces the essential DNA sequence by the positive selection marker. By homologous recombination the ES cells lose the nega- 20 CHAPTER 1. INTRODUCTION tive selection marker. Only random recombination leads to the introduction of the negative selection marker into the genome. The positive selection marker provides resistance against an antibiotic agent. For instance, the neomycin phosphotransferase gene (neo) is a gener- ally applied selection marker of E. coli origin. It provides resistance against G418 an aminoglycosidic antibiotic. On the other hand, the negative selec- tion marker, e.g. a thymidine gene (TK), makes cells vulnerable to ganciclovir treatment. Thus, the administration of G418 leads to the accumulation of recombined ES cells and the administration of ganciclovir leads to the accu- mulation of homologously recombined ES cells. However, a genomic screening (e.g. by Southern blot or PCR) needs to be applied to identify the homolo- gously recombined ES cells.

1.4 Aim of the Study

The aim of this study was to determine the physiological function of fetuin- B and to compare fetuin-B with its family member fetuin-A. Up to now the biological role of fetuin-B was unknown; the sequence homology and tissue distribution of fetuin-A and -B suggested similar but not identical functions. The main focus of this study was the analysis of the physiological function of fetuin-B in a biological system. Therefore, a fetuin-B-null mutant mouse strain was established by gene targeting in murine embryonic stem cells. The fetuin-B-deficient mice were characterized by analyzing their anatomy and morphology, by evaluating physiological parameters such as body weight, blood pressure and heart rate and by performing a clinical chemistry screen in comparison to wildtype mice. Additionally, tools were developed that would subsequently facilitate the in vitro analysis of the fetuin-B protein and gene. These tools included a recombinant fetuin-B fusion protein and reporter constructs to study fetuin- B . Chapter 2

Materials and Methods

The generation of the fetuin-B-deficient mice was a collaborations effort. The fetuin-B gene targeting vector was cloned by Dr. Bernd Denecke, IZKF Biomat, RWTH Aachen University Hospital. The ES cell transfection, blas- tocyst injection and embryo transfer were performed in the laboratory of Professor Hubert Schorle, Department of Developmental Pathology, Univer- sity of Bonn Medical School. The animals were kept in the animal facility at the RWTH Aachen University Hospital under the supervision of Dr. An- dreas Teubner. The serum chemistry was performed by Dr. Thomas Renn´e, Institute of Clinical Biochemistry and Pathobiochemistry of the University Medical Clinic, University of W¨urzburg.

Furthermore, the fetuin-B producing adenovirus was recombined and cells were infected by the adenovirus in the S2 level laboratory of Professor Ralf Weiskirchen, Institute of Clinical Chemistry and Pathobiochemistry, RWTH Aachen University Hospital.

The formulas for solutions, buffers and media are listed subsequent to each method described. Aqueous solutions were all made with distilled water. They were sterilized by autoclaving (20 min/121◦C/2 bar). Thermo-unstable ingredients were sterilized by filtration (0.2 µm) and added to the solution after autoclaving.

Commonly used molecular biology-based methods were performed follow- ing the Current Protocols in Molecular Biology [60].

21 22 CHAPTER 2. MATERIALS AND METHODS

2.1 Oligonucleotides

Lyophilized oligonucleotides were purchased from MWG Biotech AG (Ebers- berg)(0.01 µmol, HPSF purified). They were solubilized in H2O to produce 100 pmol/µl stocks.

Oligonucleotides for homologous sequence amplification:

FetuB5’Reg.3s 5’-CCGCTCGAGATGTTGGCTACTGGTTTGCTGT-3’ FetuB5’Reg.1as 5’-AGCTTTGTTTAAACCTCTAGTGAGTGGAACACAACTTCT-3’ FetuB3’Reg.1s 5’-TTGGCGCGCCTGGTTTAGTGATACGGGCCA-3’ FetuB3’Reg.1as 5’-ACGCGTCGACAAGCTGCTAGGTAGATATTTCCCAT-3’ FetuB3’Reg.2as 5’-ACGCGTCGACTGTTGAACTTTGCAAGCGACT-3’

Oligonucleotides for screening PCR and genotyping:

FetuB 8s 5’-GGGCCTGCTCAGTGTCTACC-3’ FetuB 6s 5’-CAAGTTCTAATTCCATCAGAAGC-3’ FetuB 1as 5’-GCCACCATTTCCAGAAGTCC-3’ FetuB 4as 5’-GCTTGAACGATGGGATAGGC-3’

Oligonucleotides used for fetuin-B cDNA amplification: mFetBcDNA-EcoRI-s 5’-AGAATTCTTACAGTGGAATGGGCC-3’ 2.2. GENERATING A FETUIN-B-DEFICIENT MOUSE STRAIN 23

mFetBcDNA-AgeI-as 5’-ATGACCGGTGGGTGGGAGAACCAG-3’

Oligonucleotides used for sequencing:

T7-Primer 5’-TAATACGACTCACTATAG-3’ T7mod-Primer -5’-AAATACGACTCACTATAGGG-3’ BGH-reverse 5’-TAGAAGGCACAGTCGAGG-3’

2.2 Generating a fetuin-B-deficient Mouse Strain

2.2.1 Cloning of the Gene Targeting Vector

The gene targeting vector was cloned as follows. The 5’- and 3’-homologous sequences were amplified by PCR using the Pac clone RPCIP711A0948Q2, library RPCI21, as a template. The reaction mix and the PCR program are shown in Table 2.1 and 2.2, respectively. The amplified homologous sequences were purified by using the QIAquick PCR Purification Kit (Qiagen).

Table 2.1: PCR master mix for amplifying the homologous sequences. Primers used to amplify the 5’-homologous sequence: FetuB5’Reg.3s and FetuB5’Reg.1as; the 3’-homologous sequence: FetuB3’Reg.1s and Fe- tuB3’Reg.1as; the extended 3’-homologous sequence: FetuB3’Reg.1s and Fe- tuB3’Reg.2as.

Component Final concentration Buffer (PeqLab) 1x dNTP 0.3 mM each Betain 1 M Primer s 1.2 pmol/µl Primer as 1.2 pmol/µl Taq (PeqLab) 0.04 U/µl Pwo (PeqLab) 0.004 U/µl H2O ad 50 µl Template 1 ng/µl 24 CHAPTER 2. MATERIALS AND METHODS

Table 2.2: PCR program for amplifying the homologous sequences using the MJ Research PTC200 thermocycler.

T[◦C] t Cycles 68 2 min 1x 94 1 min 94 10 sec 56 1 min 10x 68 8 min 94 10 sec 56 1 min 30x 68 8 min + 10 sec/cycle 68 24 min 1x 4 ∞

Then, the 5’-homologous sequence was digested in 1x Tango buffer with 4-fold excess of PmeI (Fermentas) at 37◦C for 4 h. Thereafter, the digestion buffer was increased to 2x Tango concentration and XhoI (Fermentas) was added. The digestion mix was incubated at 37 ◦C for 4 h. This procedure was also applied to the gene targeting vector. In parallel, the 3’homologous sequence was digested by SalI (Fermen- tas) and AscI (Fermentas) employing the enzymes 2-fold concentrated in 2x Tango buffer over night at 37◦C. The next morning, the digested PCR products and the opened gene tar- geting vector were isolated from an agarose gel (QiaQuick Gel Extraction Kit, Qiagen). The concentrations of the purified DNA fragments were esti- mated from an agarose gel before ligating the 5’-homologous sequence to the gene targeting vector. The ligation was performed over night at 16◦C using a T4 DNA ligase following the manufacturer’s instructions (Fermentas). The ligation mix was then used to transform chemically competent DH5α bacteria (Library Efficiency, Life Technologies). The resulting colonies were transferred to PCR tubes and corresponding tubes containing LB medium. The PCR tubes were heated in a microwave for 2 min before the PCR reaction mix was added. The PCR was performed according to Table 2.1 and 2.2. Positive clones were expanded immediately using the corresponding LB 2.2. GENERATING A FETUIN-B-DEFICIENT MOUSE STRAIN 25 culture. Then, a low copy plasmid midi prep (Qiafilter Plasmid Midi Kit, Qiagen) was performed to isolate the recombinant plasmid DNA. The gene targeting vector containing the 5’-homologous sequence was then digested by SalI (Fermentas) and AscI as described above and purified from an agarose gel. The purified vector’s concentration was estimated from an agarose gel, before ligating the already prepared 3’homologous sequence to the vector and transforming competent E. coli. The resulting colonies were analyzed as described above. Next, the homologous sequences were sequenced. The T7 modified se- quencing primer was used to amplify the 5’-homologous sequence and the T3 modified primer to amplify the 3’-homologous sequence. The knockout control vector was cloned equally to the gene targeting vector. Only the reverse primers that were used for the amplification of the 3’-homologous sequences were different as shown in Table 2.1.

2.2.2 Gene Targeting in Embryonic Stem Cells

Culturing Embryonic Stem Cells

The ES cells, Knut 1 (129/SvJ x C57BL/6) and BAA (C57BL/6) were thawed at day -3 and passaged 1:3 at sub confluence (day -1) onto a layer of mouse embryonal fibroblast (MEF/feeder cells). At day 0 fresh media was added to the cells, three to four hours before harvesting the cells for electroporation.

Preparation of the Targeting Vector

60 µg of the Fetuin-B knockout vector were linearized with the restriction enzyme PvuI (Fermentas) in a final volume of 3 ml. Subsequently, an aliquot was checked on a 0.7% agarose gel for complete digestion. The linearized vector DNA was purified by phenol/chloroform extraction, precipitated using 3M sodium acetate pH 5.2 and washed twice with 70% ethanol. The vector was stored in 70% ethanol at -20◦C until transfection. 26 CHAPTER 2. MATERIALS AND METHODS

Electroporation

The ethanol was removed from the linearized targeting vector and the DNA pellet was air dried before it was dissolved in PBS in a final concentration of 0.5 µg/µl. The ES cell dishes were rinsed twice with PBS (w/o Ca and Mg) and the cells were trypsinized for 5min with 1x Trypsin-EDTA (PAN). The cells were pipetted vigorously to prepare a single-cell suspension and subsequently centrifuged (250 g/5 min). The cell pellet was then resuspended in PBS (10 ml/10-cm dish) and counted. 1.2 · 107 cells were transferred to falcon tubes and pelleted by centrifugation (250 g/5 min). The cell pellet was resuspended in 750 µl RPMI w/o phenol red. 25 µg (50 µl) of the targeting vector were added to the cells. The mixture was transferred to an electroporation cuvette and placed in the electropora- tion chamber (BioRad: Gene Pulser). A single pulse (240 V/500 µF) was applied. The cells were incubated for 5 min at RT before they were resus- pended in 30 ml ES medium. The content of one cuvette was transferred to three 10-cm dishes containing feeder cells.

Positive and negative selection:

24 h after electroporation (day +1) the positive selection was started by changing the culture media to selection media containing G418 (250 µg/ml active substance). Until cryo-preservation, the selection media was replaced every day. Negative selection was started at day +5 by supplementing the selection media with ganciclovir (2 µM final concentration). Following controls were employed:

The transfection efficiency control: 1 · 103 transfected cells without se- lection media per 10-cm dish.

The Ganciclovir enrichment control: 0.5 · 106 transfected cells treated with G418.

The positive selection control: 1 · 105 non-transfected cells treated with G418. 2.2. GENERATING A FETUIN-B-DEFICIENT MOUSE STRAIN 27

Picking and Expansion of ES Cell Colonies

At day +8 three 96-well plates were prepared with feeder cells. Before start- ing the colony picking procedure the feeder cell medium was aspirated and replaced by 100 µl ES cell medium per well. A second 96-well plate was taken and filled with 35 µl of trypsin-EDTA solution. The culture dishes containing the ES cell colonies were washed twice with 5 ml PBS and then filled with 10 ml of cold PBS. A microscope was used to pick individual colonies with a 200 µl micropipettor tip in a small volume of PBS. The colonies were transferred to individual wells of the trypsin-EDTA containing dish. After finishing with one dish it was placed in the incubator (37◦C/3-5 min). Then, 50 µl of ES cell medium was added to each well. Using a multichannel pipettor, the ES cell colonies were dissociated by pipetting up and down. The content of one well was then transferred to the previously prepared feeder cell containing 96-well dishes. The cells were incubated for 2-3 days before they were pas- saged 1:3. The content of one well was transferred to three wells on three 96-well dishes. Two of these dishes were cryo-preserved while the third plate was again passaged 1:3 on three gelatinized 96 well plates and let grown until confluence. These feeder cell-free plates were used for DNA analysis.

ES cell medium: 500 ml DMEM 4.5 g/l glucose, glutamax, sodium pyruvate, Gibco 75 ml FCS ES cell tested, Hyclone 6 ml L-glutamine 100x, Gibco 6 ml non-essential amino acids Gibco 6 ml penicillin/streptomycin 100x, Gibco 1 ml Leukemia Inhibitor Factor Chemicon 1 ml 2-mercaptoethanol 50 M, Gibco

Feeder cell medium: 500 ml DMEM 4.5 g/l glucose, glutamax, sodium pyruvate, Gibco 50 ml FCS Gibco 5 ml L-glutamine 100x, Gibco 5 ml non-essential amino acids Gibco 5 ml penicillin/streptomycin 100x, Gibco 28 CHAPTER 2. MATERIALS AND METHODS

2.2.3 DNA Extraction from Cells in 96-Well Plates The ES cells were grown on gelatin-coated 96-well cell culture dishes until confluence. The cells were washed with PBS. Then 50 µl lysis buffer was added per well. The 96-well dishes were sealed with Parafilm and incubated in a humid chamber over night at 56◦C. Thereafter, the chamber was allowed to cool to RT. For DNA precipitation, 100 µl ethanol (abs.) were added to each well. Then the 96-well dishes were incubated for 2-3 h at RT. Subsequently, the DNA appeared as a filamentous precipitate that was attached to the well walls. The buffer was removed by carefully inverting the 96-well dishes. The DNA was washed twice with 80% Ethanol (100 µl/well) and air dried for 30-45 min at RT and either stored covered with 80% Ethanol at -20◦C or assessed immediately.

For DNA analysis the precipitate was eluted in 32 µl H20 by gently shaking the 96-well dishes. 3 µl of the eluted DNA were assayed in the succeeding screening PCR.

Lysis buffer: 10 mM Tris (pH 7.5) 10 mM EDTA 10 MM NaCl 0.5 % sarcosyl (w/v) Add 1 mg/ml Proteinase K just before use.

2.2.4 PCR Screening A multiplex PCR was used to screen for homologous recombined ES cells. The PCR was performed in a 96-well plate (Abgene) using 9.5 µl of the master mix and 3 µl DNA as template. The components of the master mix and the PCR program are given in Table 2.3 and 2.4, respectively. 2.2. GENERATING A FETUIN-B-DEFICIENT MOUSE STRAIN 29

Table 2.3: Screening PCR master mix.

Component Final concentration Buffer (Pharmacia) 1x dNTP 0.2 mM each Betain 1 M FetuB1as 1.2 pmol/µl FetuB6s 0.6 pmol/µl FetuB8s 1.2 pmol/µl Taq (Pharmacia) 0.04 U/µl Pwo (PeqLab) 0.004 U/µl H2O ad 25 µl Template 3 µl

Table 2.4: Screening PCR program using a MJ Research PTC200 thermo- cycler.

T[◦C] t Cycles 68 1 min 1x 94 1 min 94 10 sec 56 1 min 10x 68 8 min 94 10 sec 56 1 min 30x 68 8 min + 10 sec/cycle 68 24 min 1x 4 ∞ 30 CHAPTER 2. MATERIALS AND METHODS

2.2.5 Southern Blot

The DNA isolated from ES cell clones grown on 96-well culture dishes was transferred into a reaction tube and filled up with TE buffer to a total vol- ume of 25 µl. A master mix containing the restriction enzyme (either SspI (Fermentas) or ScaI (Fermentas)), the appropriate buffer and RNase A (Fer- mentas) were prepared. 10 µl of the master mix were added to the diluted DNA, receiving a total volume of 35 µl. The restriction was performed over night at 37◦C. The digested DNA was loaded on a 0.7% agarose gel which was run at 4 V/cm for 3.5 h. The gel was prepared for Southern blotting following the Current Protocols in Molecular Biology. The Whatman 3MM Filter Paper Wick Method was used to blot the gel. The DNA was UV-cross linked to the membrane by employing 150 mJoule (DNA side up) and 50 mJoule (DNA side down) (GS Gene Linker, Biorad). The probe was a 565 bp fragment of the knockout control vector digested with SmaI (Fermentas) and NheI (Fermentas). 25 ng were employed for radioactive labeling via α32P-dCTP integration by Exo-Klenow polymerase (Ambion) following the instructions of the Random Primed DNA Labeling Kit (DECAprimeTMII, Ambion). The membrane was pre-hybridized for 2 h with hybridization buffer (Ambion) at 43◦C. Hybridization was performed over night. Following this, the membrane was washed with prewarmed 2x SSC/0.1%SDS (2x 30 min at 43◦C) and then 0.1x SSX/0.1% SDS (3x 30 min 43◦C). Subsequently the membrane was enclosed by cera wrap and put into a film cassette. The exposition time was 48 h.

2.2.6 Blastocyst Injection

Homologous recombined ES cell clones (Knut 1 and BAA) were expanded (4th passage) and then injected into blastocysts derived from superovulated Balb/c female mice as described in Manipulating the Mouse Embryo.

2.2.7 Embryo Transfer

An embryo transfer was performed in order to transfer fetuin-B-deficient mice into the specific pathogen free (SPF) barrier of the animal facility. Therefore, homozygous fetuin-B-deficient males were mated with wildtype C57BL/6J females. The two-cell stage embryos were then implanted into the oviduct 2.2. GENERATING A FETUIN-B-DEFICIENT MOUSE STRAIN 31 of pseudo-pregnant SPF C57BL/6J recipient females. The procedure was performed as described in Manipulating the Mouse Embryo.

2.2.8 Animal Husbandry and Breeding

The mice were kept in the university hospital animal facility (Institut f¨ur Versuchstierkunde, RWTH Aachen) under standardized environmental con- ditions and a 12/12 h light/dark period. The animals were fed with standard rodent food (Altromin Nr. 1324) and water ad libitum. Heterozygous fetuin-B-deficient mice with mixed genetic background were continuously mated with mice of the inbred strains C57BL/6J or DBA/2, in order to generate two mice strains deficient for fetuin-B with different but defined genetic backgrounds. Ten generations of backcrossing leads to a genetic background that resembles 99,9% of the inbred genetic background. The mating system was either based on monogamous pairs (one male and one female per cage) or trios (one male and two females per cage), employing either heterozygous males and wildtype females or wildtype males and heterozygous females depending on their availability.

2.2.9 Genotyping

Isolating DNA from Tail Biopsies

Genomic DNA of tail biopsies (± 0.5 cm) of ear marked individuals was isolated by Proteinase K digestion and isopropanol precipitation following the protocol of Laird et al. The isolated DNA was dissolved in 200 µl TE buffer and stored at 4◦C.

Proteinase K digestion mix: 100 mM Tris/HCl pH8.5 5 mM EDTA 0,2% SDS 200 mM NaCl 100 µg/ml Proteinase K 0.5 ml total volume/sample 32 CHAPTER 2. MATERIALS AND METHODS

Genotyping PCR

A multiplex PCR was used to screen for wildtype and fetuin-B-deficient alleles. The PCR was performed in PCR stripes employing 25 µl of the master mix (Table 2.5) and 1 µl of the tail DNA. The PCR program is given in Table 2.6.

Table 2.5: Genotyping PCR master mix.

Component Final concentration Buffer (Promega) 1x dNTP 0.2 mM each Betain 1 M FetuB4as 1.2 pmol/µl FetuB6s 0.6 pmol/µl FetuB8s 1.2 pmol/µl Taq (Promega) 0.025 U/µl H2O ad 25 µl Template 1 µl

Table 2.6: Genotyping PCR program using a MJ Research PTC200 ther- mocycler.

T[◦C] t Cycles 94 2 min 94 10 sec 56 30 sec 24x 68 30 sec 68 7 min 4 ∞

2.3 Analysis of fetuin-B-deficient Mice

2.3.1 Western Blot Wildtype and fetuin-B-deficient mouse serum was analyzed by loading 0.1 µl serum in 10 µl 1x Laemmli loading buffer on a 10% PAA gel. The protein gels were then blotted onto a nitrocellulose membrane (Protran, Whatman, Schleicher und Schuell) at 1.5 mA/cm2 for 45 min, using a semi-dry blotting 2.3. ANALYSIS OF FETUIN-B-DEFICIENT MICE 33 device (Biorad). The transfer was evaluated by Ponceau-S staining. The membrane was blocked for 30 min at 37◦C with blocking solution. First and second antibody were diluted in blocking solution (for antibody concentra- tions see Table 2.7) and incubated for 45 min at 37◦C. After each antibody incubation the blot was rinsed 3 x 5 min in washing solution. Protein detec- tion was achieved by chemiluminescence.

Blocking solution: Washing solution: 1x PBS 1x PBS 0.05% Tween 20 0.05% Tween 20 5% dried milk powder

Table 2.7: Antibodies and concentrations used for Western blot analysis.

First antibody Derived from Dilution Second antibody Dilution α-mouse fetuin-A rabbit K98 1:5000 α-rabbit HRP 1:5000 α-mouse fetuin-B rabbit K319 1:4000 α-rabbit HRP 1:5000

2.3.2 Immunohistochemistry The immunohistochemistry was performed on cryo-sections. The sections were kept at -80◦C over night. Thereafter, the sections were incubated in fixative at -20◦C for 5 min, and washed 2x in washing buffer (5 min each). Next, the fixed sections were perfused by incubating them in perfusion buffer at RT for 10 min. Then, the sections were again rinsed with washing buffer and blocked with absorption buffer at RT for 30 min. The primary antibody α-mouse fetuin-B (Table 2.7) was diluted in absorp- tion buffer (1:300), spotted onto the sections and then incubated at RT for 2 h in a humidified chamber. The sections were washed 4x in washing buffer (5 min each) before the secondary antibody ALEXA Fluor 546 goat anti-rabbit IgG ((H+L), Molecular Probes) was applied (1:500 in absorption buffer) at RT for 1 h. Thereafter, the sections were again rinsed 4x in washing buffer. Subsequently, the sections were carefully dried using paper tissue and three drops of Immu-Mount (Thermo Shandon) were put onto the sections 34 CHAPTER 2. MATERIALS AND METHODS in order to cover them with a cover glass. After 30 min, the sections were sealed with clear nail polish.

Fixative: Washing buffer: 50% methanol 1x PBS 50% acetone 0.05% Tween 20

Absorption buffer: Perfusion buffer: 1x PBS 1x PBS 0.05% Tween 20 0.1% Tween 20 10% FCS % Tween 20

2.3.3 Collecting Mouse Oocytes

In order to collect newly ovulated oocytes, wildtype and fetuin-B-deficient females were mated with wildtype males. As indicator that mating had occurred, females were checked for vaginal plug formation in the morning. Plug positive females were subsequently sacrificed by cervical disloca- tion. The oocytes were isolated from the ampulla, the upper part of the oviduct. Since ovulated oocytes are surrounded by a cloud of cumulus cells, the collected cumulus was incubated in M2 medium containing 0.3 mg/ml hyaluronidase (Sigma) for 2-3 min to set the oocytes free. Then, the oocytes were washed in several drops of M2 medium and further cultivated in a KSOM microdrop culture under a layer of embryo-tested mineral oil (Sigma) in an cell culture incubator. A comprehensive protocol describing the dissection of the oviduct, the isolation of oocytes from the latter and the cultivation of the oocytes is detailed in Manipulating the Mouse Embryo [44]. Micrographs of the cultured oocytes were taken daily using light mi- croscopy (Leitz DMIRB, Leica) with or without differential interference op- tics. For the latter, the oocytes needed to be cultivated on glass culture dishes. The pictures were analyzed and archived with the DISKUS software (Karl Hilgers, K¨onigswinter). 2.3. ANALYSIS OF FETUIN-B-DEFICIENT MICE 35

2.3.4 Primary Morphological Analysis

The primary morphological analysis was performed according to the screen- ing protocol in Standards of Mouse Model Phenotyping which is a modified protocol based on Fuchs et al. [61] The protocol includes observing the mice:

• in a cage

• on the experimentator’ hand

• holding them by the neck

• suspending them by the tail

• dropping them into the cage

12 week old animals were examined. Additionally, when handling the animals in general, a special emphasis was made on observing them according to the protocol.

X-Ray Analysis

For X-ray analysis six and 12 months old male and female mice were anes- thetized for a short-term with isofluran. The X-ray pictures were taken in the ”Klinik f¨urRadiologische Diagnostik, UK Aachen” using a mammography machine (Senographe DMR X-ray System, GE Medical Systems SA) with the following exposition parameters: 25 kV, 35 mAs, and 1.9 x magnification.

2.3.5 Necropsy

The necropsy was performed following the instructions published in ”The ”. Prior to collecting the organs, the mice where perfused by injecting PBS into the left ventricle of the heart and incising the right ventricle of the heart for blood flow. Organs and tissue were fixed with Bouin’s solution over night, washed excessively with tab water and stored in 70 % Ethanol. 36 CHAPTER 2. MATERIALS AND METHODS

Bouin’s solution: 750 ml sat. ag. picric acid 50 ml glacial acetic acid 250 ml 37% formalin

2.3.6 Histology The Bouin’s fixed organs were dehydrated using a dehydration apparatus (Tissue-Tek VIP, Sakura):

70% ethanol 2x 1 h 96% ethanol 2x 1 h 100% ethanol 3x 1 h Xylol 3x 1 h Paraffine, 60◦C 4x 1h

Subsequently, the organs were imbedded in paraffin and cut into 4 µm sections after cooling. The sections were transferred onto glass slides and in- cubated over night at 60◦C. The next morning, the sections were rehydrated:

Xylol 3x 3 min 100% ethanol 2x 3 min 96% ethanol 3 min 80% ethanol rinsed 70% ethanol rinsed

H2O rinsed

After that, the sections were haematoxylin and eosin stained and imme- diately dehydrated:

Xylol 3x 3 min Heamatoxilin 7 min

H2O rinsed Tab water, lukewarm 10 min Eosin 10 min 80% ethanol rinsed 2.3. ANALYSIS OF FETUIN-B-DEFICIENT MICE 37

100% ethanol 2x 2 min Xylol 2x rinsed

Then, one drop of Vitroclud (Langenbrinck, Teningen) was applied in order to cover the sections with a cover glass.

2.3.7 Weighing

In order to follow up the animals’ weight corresponding to their genotype, three week old F2-generation animals containing all three genotypes were ear marked, genotyped and then weighed once a week.

2.3.8 Blood Pressure Measurement

The Blood Pressure Analysis System BP-2000 (Visitech Systems) was used to determine the mice’s blood pressure and pulse rate. One measuring ses- sion comprised ten habitation measurements and ten subsequent recorded measurements of systolic/diastolic blood pressure and pulse rate. One ses- sion was performed per day. The first five sessions were training phases only. The results of the next five sessions were used for blood pressure and pulse rate determination.

2.3.9 Glucose Tolerance Test

The glucose tolerance test was performed following the instructions of the Joslin Diabetes Center (http://www.whitelabs.org) The mice were transferred to a new cage with water but without food and fasted for 15 h over night prior to the glucose tolerance test. The next morning, the animals were weight and four animals at a time were fixed on the Blood Pressure Analysis System BP-2000 platform (Visitech Systems), which they were already adapted to. In order to determine the baseline blood glucose, the tail was nicked with a scalpel by a horizontal cut of the very end and a drop of blood was ap- plied to a sensor of the Ascensia Elite glucometer (Bayer). After that, the animals were challenged by injecting 2 g/kg body weight of 20 % D-glucose intraperitoneally with an insulin syringe and put back into their cage. 38 CHAPTER 2. MATERIALS AND METHODS

15, 30, 60 and 120 min post glucose injection, the mice were again fixed on the blood-pressure platform and the blood glucose was measured by sampling blood from the nicked tail of each mouse.

2.3.10 Clinical Chemical Screening Serum Analysis

Blood was collected by cardiac puncture after an overdoses of isofluran as detailed in ”The Laboratory Mouse”. The blood was applied to a serum tube and incubated for 1 h at RT. Then, the serum tube was centrifuged (1000 g/10 min) and the serum supernatant was transferred to a reaction tube. The serum was cryo-preserved at -80◦C.

2.4 Recombinant Expression and Purification

2.4.1 Cloning Murine Fetuin-B PCR Amplification of Murine Fetuin-B cDNA

The fetuin-B cDNA sequence was amplified by PCR using the murine cDNA clone MGC:25848 (BC018341) as a template. Table 2.8 shows the reaction components. Mix I and II were added 1:1 on ice. Table 2.9 shows the PCR program. The resulting blunt-ended PCR product was purified by agarose gel extraction (QiaQuick Gel Extraction Kit, Qiagen).

Cloning into pGEM-T

An A-tailing procedure was applied to the purified PCR product: 5 µl of the purified PCR product plus 1 µl of Taq DNA Polymerase 10x Reaction

Buffer with MgCl2 plus dATP in a final concentration of 0.2 mM plus 5 units of Taq DNA Polymerase plus water to a final volume of 10 µl were mixed and incubated at 70◦C for 30 min. Of this, 2 µl were employed in the ligation reaction with Promega’s pGEM-T vector as detailed in the pGEM-T technical manual. The transformation of the ligation reaction was performed according to the pGEM-T technical manual. 2 µl of the ligation reaction were employed using high efficiency JM109 competent cells (Promega). 2.4. RECOMBINANT EXPRESSION AND PURIFICATION 39

Table 2.8: Murine Fetuin-B cDNA Amplification. PCR reaction mix.

Mix I Component final concentration dNTP 0.6 mM each mFetBcDNA-EcoRI-s 0.3 pmol/µl mFetBcDNA-AgeI-as 0.3 pmol/µl Template 30 pg/µl H2O ad 50 µl

Mix II Component final concentration buffer (PeqLab) 1x Pwo (PeqLab) 0.004 U/µl H2O ad 50 µl

Table 2.9: PCR program for amplifying murine fetuin-B cDNA using a MJ Research PTC200 thermocycler.

T[◦C] t Cycles 95 2 min 95 15 sec 55 30 sec 72 2 min 10 95 15 sec 55 30 sec 72 2 min 30 + 20 sec/cycle 95 15 sec 72 7 min 4 ∞ 40 CHAPTER 2. MATERIALS AND METHODS

The resulting colonies were transferred to PCR tubes and corresponding tubes containing LB medium. The PCR tubes were heated in a microwave for 2 min before the PCR reaction mix (Table 2.8) was added. The PCR was performed according to Table 2.9. Positive clones were expanded immediately using the corresponding LB culture. Then, a plasmid midi prep (Qiafilter Plasmid Midi Kit, Qiagen) was performed to isolate the recombinant plasmid DNA.

Sub-Cloning into pcDNA3.1-V5-His

The purified recombinant plasmid was digested by EcoRI/AgeI (Fermentas). The excised PCR product was purified from an agarose gel (QiaQuick, Qi- agen). The concentration of the purified PCR product was estimated from an agarose gel before it was ligated into the EcoRI/AgeI opened eukaryotic expression vector pcDNA3.1-V5-His (Invitrogen). For the ligation reaction 2 µl of the prepared vector, 2 µl 10x Ligase Buffer, 1 µl Ligase (Fermentas), 5 µl water and 10 µl of the purified PCR product were applied. The transformation of the ligation and the analysis of the resulting colonies was performed as described above. Additionally the purified recombinant plasmid DNA was digested by BamHI and also XhoI (Fermentas). Two positive clones were sequenced using the Applied Biosystems cycle sequencing technology at the Fraunhofer IME (Aachen) using an ABI PRISM 3730 Genetic Analyzer. The Sequences were analyzed using Staden Package software.

Sub-Cloning into the Adenoviral Shuttle Vector

The sequenced recombinant plasmid was digested by MfeI/DraIII (Fermen- tas). The 2.5 kb fragment was purified from an agarose gel (QiaQuick, Qi- agen) and the overhanging ends were either filled or digested by a T4 Poly- merase reaction:

28 µl purified fragment 8 µl 5X T4 Polymerase Buffer 2 µl 2 mM dNTP mix 1 µl T4 Polymerase 11◦C for 20 min + 70◦C 10 min 2.4. RECOMBINANT EXPRESSION AND PURIFICATION 41

The blunt ended fragment was then purified (QiaQuick, PCR Purification Kit, Qiagen) and its remaining concentration was estimated from an agarose gel before it was ligated to the EcoRV (Fermentas) opened adenoviral shuttle vector, p∆E1sp1A. The ligation, transformation and analysis of the resulting colonies were performed as described above. Additionally the resulting shuttle vector was digested by BamHI and also HindIII (Fermentas).

2.4.2 Generating a Fetuin-B Transfecting Adenovirus

The construction of replication-deficient recombinant adenoviruses (Ad5) was based on the plasmids p∆E1sp1A and pJM17 (Biosystems Inc., Toronto, Canada) and their homologous recombination in 293 cells. The protocol is detailed in Weiskirchen et al. [62].

2.4.3 Expression and Purification of Fetuin-B

Recombinant Expression of Fetuin-B

The virus containing supernatant of 293 cells was obtained and added to COS-7 cells. After an over night incubation period the media was changed to a serum-free culture and incubated for another 24 h before obtaining the cell culture supernatant in order to purify the expressed fetuin-B protein.

Purification of Recombinant Fetuin-B

The cell culture supernatant of fetuin-B expressing COS-7 cells was collected and filtered through a 0.45 µm filter. The His-tagged protein was purified using the AKTAdesign¨ liquid chromatography system applying 1 ml HisTrap affinity columns from GE Healthcare following the manufacturer’s instruc- tions. The HisTrap columns were stripped and recharged with 0.1 M NiCl2 after every run also following the manufacturer’s instructions. The fractions containing the purified proteins were pooled and dialyzed in 1 l PBS for 2 h and 4 l PBS over night. 42 CHAPTER 2. MATERIALS AND METHODS

Binding buffer: Elution buffer: 20 mM sodium phosphate 20 mM sodium phosphate 0.5 M NaCl 0.5 M NaCl 20 mM imidazole 500 mM imidazole pH7.4 pH7.4

Sequencing the Purified Fetuin-B

The protein sequencing was performed at the TOPLAB (Gesellschaft f¨ur angewandte Biotechnologie mbH, Martinsried, Germany) using an Applied Biosystems Procise Sequencer. 10 pM standard and recombinant murine fetuin-B each were applied at a sampling rate of 4.0 Hertz. The data were analyzed using Smooth Degree 9, Interpolated Baseline with the following settings: analysis limits 1 - 128; integration and baseline limits 3.20 - 19.00 min and 0.10 - 1.00 min., respectively; peak half with and peak separation 0.05 min; search length 0.10 min; RT factor 0.80 and sensitivity 1.00.

2.5 In Vitro Analysis of Fetuin-B

2.5.1 Precipitation Inhibition Assay

The calcium phosphate precipitate inhibition assay is based on the measure- ment of radioactively labeled calcium (45Ca2+) in a saturated calcium and phosphate solution, that incorporates into the growing calcium phosphate precipitate [63]. Thereby, the inhibitory potential of a test protein is directly correlated to the measured amount of the labeled precipitate, at the end of the experiment. The protein and serum samples were incubated with the buffered precip- itation mix as triplicates for 90 min at 37◦C. After the incubation period the precipitate was pelleted by centrifugation (5 min./10,000 g). The pellet was then solubilized in 200 µl of 1 % acetic acid and subsequently mixed with 1 ml of szintillation buffer. The measurement of the beta radiation was measured using the beta counter Wallac 1450 MicroBeta TriLuxt. 2.5. IN VITRO ANALYSIS OF FETUIN-B 43

Precipitation reaction mix:

5 mM CaCl2

3 mM NaHPO4 50 mM Tris pH7.4 140 mM NaCl final volume of 200 µl

2.5.2 Mink Lung Cell Reporter Assay

Mink lung epithelial cells (MLEC) stably transfected with a PAI-1/Luciferase reporter were cultivated in geneticin medium and passaged 1:10 every three days. For an overview see Abe et al. [64]. For the TGF-β assay, the cells were trypsinized at 37◦C for 4 min, sus- pended in 8 ml of normal medium and pelleted by centrifugation (10 min/1700 g/4◦C). Then, the supernatant was removed and the cells were resuspended in 10 ml normal medium. 16000 cells/100 µl were plated in each well of a 96-well cell culture dish. Twelve hours later, the media was removed from the cells and 100 µl assay medium were applied per well in triplicates. After an incubation period of 20 h, the cells were removed from the incubator and cooled to room temperature for 15 min. Then, 100 µl of reconstituted Steady-Glo Luciferase Assay Buffer (Promega) were applied to each well. After careful shaking for 6 min at RT, 175 µl of the supernatant were transferred to a white 96-well micro-titer dish and analyzed using a beta counter Wallac 1450 MicroBeta TriLuxt.

Geneticin medium: Normal medium: DMEM DMEM inc. glutamine and glucose inc. glutamine and glucose 10% FCS 10% FCS 1% penicillin/streptamycin 1% penicillin/streptamycin 200 µg/mlG418 44 CHAPTER 2. MATERIALS AND METHODS

Assay medium: DMEM inc. glutamine and glucose 0.1% BSA 1% penicillin/streptamycin

2.6 Promoter Analysis

2.6.1 Cloning of Reporter Constructs

The potential fetuin-B promoter sequences were amplified by PCR employing murine genomic DNA as a template (see Tables 2.10 and 2.11). The resulting PCR products were purified (Qiaquick, PCR Purification Kit, Qiagen) and subsequently digested by SmaI (Fermentas) for 3 h at 37◦C. After that, the DNA fragments were again purified and digested by KpnI (Fermentas) over night at 37◦C. The vector pEGFP-1 (Invitrogen) was also digested as described above. The DNA fragments were purified and their concentration was estimated from an agarose gel. The ligation was performed with a T4 Polymerase (Fermentas) following the manufacturer’s instructions at 16◦C over night. The transformation of chemically competent E.coli (DH5-α) was per- formed by heat shock. The resulting colonies were checked by restriction analysis (BamHI, XcmI) of plasmid mini prep DNA. Positive clones were grown in LB medium, thereof, the plasmid midi prep DNA was used for sequencing.

2.6.2 Transfection of Reporter Constructs

The reporter constructs were transfected by Lipofectamine 2000 (Invitrogen) following the manufacturer’s instructions. 16 h prior to transfection 0.5-2·105 cells were seeded to 24-well cell culture dishes. The media contained FCS but no antibiotics. 0.8 µg DNA and 2.5 µl Lipofectamine 200 were diluted in 50 µl medium without FCS, each. Then, both solutions were mixed and incubated for 2.6. PROMOTER ANALYSIS 45

Table 2.10: Amplification of promoter sequences PCR master mix. The following primers were used: mFetuBProm1-s and -as for the amplification of the first possible murine fetuin-B promoter region; mFetuBProm2.1-s and -as for the amplification of the second possible promoter region.

Component Final concentration Buffer (PeqLab) 1x dNTP 0.2 mM each Primer s 0.6 pmol/µl Primer as 0.6 pmol/µl Pwo (PeqLab) 0.025 U/µl H2O ad 50 µl Template 0.2 µg

Table 2.11: PCR program for amplifying promoter sequences using a MJ Research PTC200 thermocycler.

T[◦C] t Cycles 68 2 min 1x 94 1 min 94 10 sec 60 1 min 35x 68 1 min 68 8 min 4 ∞

20 min before applying the resulting 100 µl to the cells. After 5 h 10% FCS were added to the media. The promoter activity was assayed by fluorescence microscopy 24 h after the transfection.

Chapter 3

Results

3.1 Generating a Fetuin-B-deficient Mouse Strain

3.1.1 Gene Deletion and the Targeting Vector

A mouse-genomic PAC clone library (129/SvevTACfBR, RZPD, Germany) was screened for the genomic Fetub sequence. A fetuin-B cDNA fragment consisting of exons 2-8 (834 bp) was used as a probe. The identified Fetub positive PAC clone (RPCIP711A0948Q2 library RPCI21) also contained the entire gene for fetuin-A (Ahsg). This confirmed that the genes for fetuin- A and fetuin-B are located in the same chromosome region (16 B1) in the mouse genome, separated by 18.8 kb [65] and ensured that the PAC clone contained the entire 5’-region of the Fetub gene. The strategy for disrupting the Fetub gene was based on the pGKneo- TV gene targeting vector, which had already been used in our group for the targeted deletion of the Ahsg and HRG genes [25, 66]. The vector con- tains a PGKneo-cassette, a gene employing resistance to neomycine, which is necessary for the positive selection of transfected ES cell clones. The PGKneo-cassette is flanked by a multiple cloning site on each side. These enable the insertion of 5’- and 3’- homologous regions alongside the region which is supposed to be replaced by the neo-cassette. Further features are two thymidine kinase genes (TK) that are located upstream from the first multiple cloning site and downstream from the second multiple cloning site. Only in case of a non-homologous recombination would these genes be in- serted into the ES cell genome, making the cells sensitive to Ganciclovir.

47 48 CHAPTER 3. RESULTS

Figure 3.1 gives a graphical overview of the gene targeting vector and the gene deletion strategy.

FetuB 5R3s 5R1as ATG 3R1s 3R1as

Fetuin-A E1 E2 E3 E4 E5 E6 E7 E8 E 7 5'-homology 3'-homology XhoI PmeI AscI SalI

neo

ko-vector TK (pGKneo-TV) TK

Figure 3.1: The genomic fetuin-B sequence and the fetuin-B gene targeting vector are shown. The exons (E1-E8) and the amplified homologous sequences are highlighted by black boxes. The primers used for the amplification of the homologous regions are shown with red arrows. Note that the translation start codon (ATG) will be deleted by the homologous recombination of the gene targeting vector with the genomic DNA.

Using the fetuin-B positive PAC clone as a template, a 2.7 kb 5’ homolo- gous region located between exon 1 and exon 2 was amplified by PCR. The sense primer (FetuB5R3s) used contained an internal XhoI restriction site, the anti-sense primer (FetuB5R1as) a PmeI restriction site. This permitted the directed insertion of the 5’-homologous sequence into the multiple cloning site of the pGK-neo-Tv vector upstream of the neo-cassette using the unique restriction sites for XhoI and PmeI. The 3’-homologous region, located be- tween exon 3 and exon 5, was also amplified by PCR using a sense primer (FetuB3R1s) containing an AscI restriction site and an anti-sense primer (FetuB3R1as) containing a SalI restriction site. The insertion of the 3’ ho- mologous region was performed using the unique AscI and SalI restriction sites located in the multiple cloning site downstream from the neo-cassette as shown in Figure 3.1. Using the 5’-homologous region located between exon 1 and 2 and the 3’ homologous region between exon 3 and 5 means replacement of exon 2 and 3 by the neo-cassette. Exon 2 contains the translation-start 3.1. GENERATING A FETUIN-B-DEFICIENT MOUSE STRAIN 49 codon, that determines the ORF of the fetuin-B gene. Replacing this codon destroys the functionality of the fetuin-B gene.

Knockout Control Vector

A knockout control vector was designed to model the recombinant fetuin-B genetic locus. For the establishment of the screening procedure, the knock- out control vector was used as a positive control resembling the homolo- gously recombinated fetuin-B allele (Figure 3.2). A 3’-homologous sequence that was extended towards the 3’-end was amplified, exhibiting the primer- and probe-binding sites that were required for the screening for homologous recombination.

5R3s 5R1as 3R1s 3R1as 3R2as A fetuin A E1 E2 E3 E4 E5 E6 E7 E8 E 7 5'-homology 3'-homology B TK neo TK

3'-homology extended C TK neo TK

D neo fetuin-A E1 -235 bp E4 E5 E6 E7 E8 E 7

Figure 3.2: Gene targeting and knockout control vector. The genomic fetuin- B sequence and relevant features of the gene targeting vector as well as knock- out control vector are shown: (A) genomic Fetub sequence, (B) gene targeting vector, (C) knockout control vector, (D) recombinant Fetub locus. Exons are depicted by black boxes. The homologous sequences are red colored. The primers used for amplifying the homologous sequences of the gene targeting and the knockout control vector are shown by red arrows. TK: thymidine kinase; neo: neomycin resistance.

The pGKneo-Tv vector containing the 5’-homologous region as described above was used to insert the extended 3’-homologous sequence which was amplified with the same sense primer (FetuB3R1s) as the 3’-homologous sequence inserted into the gene deletion vector but with a different anti- 50 CHAPTER 3. RESULTS sense primer (FetuB3R2as). The extended 3’-homologous sequence was also inserted into the vector using the AscI and SalI restriction sites downstream from the PGKneo-cassette.

3.1.2 Establishing the Screening Methods

PCR Screening To identify homologously recombined ES cells, a screen- ing strategy was developed. I decided to analyze the transfected ES cells by using a multiplex PCR, that could amplify the wildtype fetuin-B allele and the homologously recombined fetuin-B locus simultaneously (see Figure 3.3).

P8 P1 A Fetuin-A E1 E2 E3 E4 E5 E6 E7 E8 E 7 P6 5'-homology 3'-homology B TK neo TK

P6 P1

C TK neo TK

WT PCR product 3,4 kb

KO PCR product 2,5 kb

Figure 3.3: Fetuin-B knockout PCR screening strategy. (A) genomic FetuB sequence, (B) gene targeting vector, (C) knockout control vector. The primer binding sites used for the screening PCR are indicated by red arrows (P1: FetBgenscreen1as; P6: FetBgenscreen6s; P8: FetBgenscreen8s). The wildtype (WT) and knockout (KO) specific PCR products are revealed by red bars.

One anti-sense primer (P1), whose binding site is not present on the gene deletion vector, and two different sense primers were used for the multiplex PCR. One of the sense primers was wildtype specific (P8), while the other primer was specific to the neo-cassette (P6). The primer binding sites where chosen such that the wildtype PCR product at 3.4 kb was larger than the homologously recombined fetuin-B locus at 2.5 kb, making the latter product the more favorably amplified PCR product in the multiplex PCR. The primers were assayed for specificity using mouse-genomic DNA, for the wild type allele (Figure 3.4 lanes 2-4), and the knockout control construct 3.1. GENERATING A FETUIN-B-DEFICIENT MOUSE STRAIN 51

Figure 3.4: Establishing the knockout screening PCR: Gel electrophore- sis picture showing the specificity of the designed screening primers and the sensitivity of the screening system. 1AS: anti-sense primer; 6S: sense primer, specific for knockout sequence; 8S: sense primer, specific for wildtype sequence.

(lanes 8-10) as templates. The latter was employed because of its resemblance to the homologously recombined fetuin-B allele. To simulate the screening procedure, the knockout control vector was added to the genomic DNA in an equimolar concentration (lanes 15, 17, 19). The sensitivity of the multiplex PCR to detect even little amounts of DNA was tested by adding a decreasing amount of template to the PCR. Figure 3.4 shows that the primers are specific for the amplification of the 3.4 kb wildtype and 2.5 kb knockout sequence as shown in Figure 3.3. Additionally, there is an unspecific PCR product (about 1 kb in size) using the wildtype primers (8S/1AS). However, there is no loss of specificity from 3 µg to 0.025 µg of DNA.

Verifying by Means of Southern Blot A genomic sequence analysis of the targeted FetuB gene revealed the loss of an SspI and ScaI restriction site and the addition of a ScaI restriction site due to the insertion of the PGKneo-cassette. This is shown in Figure 3.5. The SspI wildtype DNA fragment is 4 kb in size and smaller than the knockout DNA fragment (6.5 kb) due to the loss of an SspI restriction site. 52 CHAPTER 3. RESULTS

SspI SspI ScaI ScaI SspI

A fetuin-A 5'-homology 3'-homology probe E 7

B TK neo TK

C TK neo TK probe SspI KO fragment: 6475 bp SspI WT fragment: 3990 bp

ScaI WT fragment: 3704 bp ScaI KO fragment: 3380 bp

Figure 3.5: Southern Blot screening strategy. (A) genomic FetuB sequence, (B) gene targeting vector, (C) knockout control vector. Blue arrows indicate SspI, green arrows ScaI restriction sites; the orange bar resembles the probe at its binding site.

When looking at the ScaI restriction pattern, the wildtype DNA fragment is 3.7 kb in size and larger than the knockout DNA fragment (3.4 kb), due to the additional ScaI restriction site that is located within the neo-cassette. A probe was made by digesting the knockout control vector with the restriction enzymes SmaI and NheI. A 565 bp DNA fragment of the 3’- end of the 3’-homologous sequence was purified and analytical amounts were analyzed by digesting with XcmI. The resulting bands (399 bp and 166 bp) verified the purified DNA band as the correct 565 bp probe. The screening of targeted ES cell DNA was simulated by employing mouse genomic DNA and the knockout control vector which was added to the ge- nomic DNA in equimolar amounts. The predicted DNA fragments were detected in the range 5 µg to 0.5 µg by autoradiography of the Southern blots for 74 h. This is shown in Figure 3.6.

3.1.3 Gene Targeting in Embryonic Stem Cells

30 µg of the linearized gene targeting vector was required for each transfec- tion of Knut 1 ES cells (129/SvJ x C57BL/6J genetic background) and BAA 3.1. GENERATING A FETUIN-B-DEFICIENT MOUSE STRAIN 53

ScaI SspI WT KOC mixed WT KOC mixed

6,5 kb

4 kb 3,7 kb 3,4 kb

Figure 3.6: Establishing the Southern blot screening: Wildtype genomic (WT) and knockout control vector (KOC) DNA treated with ScaI (left side) and SspI (right side). Mixed: genomic DNA and equimolar masses of the knockout control vector resembling 5 µg, 1 µg, 0.5 µg and 0.1 µg.

ES cells (C57Bl/6J genetic Background). Two transfections were performed for each cell type. Therefore 300 µg of the knockout vector were linearized with the restriction enzyme PvuI and purified by phenol-chloroform extrac- tion. Analytical amounts of the linearized knockout vector were checked for complete linearization on an agarose gel as shown in figure 10.7. The re- maining DNA concentration was measured (0.1 µg/µl) and 6 x 300 µl were precipitated using sodium acetate precipitation.

After G418 selection and ganciclovir enrichment, 262 Knut 1 and 288 BAA resistant ES cell colonies were transferred to 96-well plates. The cells were grown to confluency and passaged to four replicate 96-well plates. One of the replicate plates was cryo-preserved for eventual expansion of homologously recombined ES cell clones. The other plates were used for the following screening procedure. 54 CHAPTER 3. RESULTS

3.1.4 Screening for Recombinant ES Cell Clones

PCR Screening: The DNA extraction was performed for one of four repli- cate 96-well dishes containing the 262 expanded G418 resistant Knut 1 ES cell colonies and the 288 BAA ES cell colonies. The remaining dishes were kept cryo-preserved for later analysis. The purified genomic DNA was added to the previously established screening PCR. To assess the accuracy of the PCR analysis, an equimolar concentration of genomic DNA and knockout control vector was used as a positive control, simulating the wildtype and the homologously recombined fetuin-B allele. Figure 3.7 shows a representative result of the PCR screening. Colony A3 (Knut 1) and H7 (BAA) carry the fetuin-B wildtype and knockout allele which are 3.4 kb and 2.5 kb of size, respectively. The homologous recombina- tion rate was one in 262 for the resistant Knut 1 ES cells and one in 288 for the resistant BAA ES cells. Taking into account that the initial cell number was 2.4 · 107 for both ES cell lines, the total homologous recombination rate was 4.16 · 10−8.

Figure 3.7: Screening PCR showing positive ES cell clones: The PCR prod- ucts of the positive control (PK) and the two ES cell colonies with heterozygous fetuin-B loci (A3 and H7) are marked with a star.

Verifying by Means of Southern Blot: The PCR screening result was further verified by a Southern blot as described in section ”Establishing the fetuin-B Knockout Screening Methods” above. Figure 3.8 shows the Southern Blot. It displays the expected radio-labeled bands at 6.5 kb and 4 kb for the SspI treated, as well as 3.7 and 3.4 kb for the ScaI treated genomic DNA of colony A3 (Knut 1) and H7 (BAA). 3.1. GENERATING A FETUIN-B-DEFICIENT MOUSE STRAIN 55

Figure 3.8: Verifying homologously recombined ES cells by Southern blot: 5 µg and 0.5 µg of genomic DNA and equimolar masses of the knockout-control vector treated with ScaI and SspI were used as positive controls. Colonies 1G7 and 3B10, both tested negative for homologous recombination by PCR, were employed as negative controls.

Summarizing the results: The targeted deletion of the fetuin-B gene in the two ES cell lines Knut 1 and BAA, resulted in one ES cell clone each, tested positive for homologous recombination by PCR and Southern Blot.

3.1.5 Blastocyst Injection and Chimera

The two ES cell clones A3 (Knut 1) and H7 (BAA) where expanded and then injected into blastocysts derived from superovulated Balb/c female mice. Next, the injected blastocysts were transferred by uterus transfer into pseudo- pregnant foster mothers. The data summarizing the cell injection and embryo transfer experiments are given in Table 3.1. Of 95 transferred blastocysts, one chimera containing BAA ES cells was derived. The chimera exhibited 50 % chimerism but did not pass on the mutation through the germ line. Even after five attempts no offspring derived from the manipulated stem cells was delivered. The manipulated Knut 1 ES cells were more productive. Two chimeras, 56 CHAPTER 3. RESULTS

Table 3.1: Blastocyst injection and outcome

] of ES cell transferred pups chimera/ germ line donors clone blastocysts % chimerism transmission 10x A3 10 6 1 male/90% 100% Balb/c A3 10 2 1 male/80% 100% A3 10 4 - A3 10 - -

10x H7 10 4 1 male/50% 0 Balb/c H7 10 6 - H7 10 3 - H7 10 2 -

10x H7 11 1 - Balb/c H7 11 - H7 11 - H7 11 - H7 11 -

showing 80 and 90 % chimerism, were obtained from 40 transferred blasto- cysts. In both chimeras the Knut 1 stem cells had contributed to the germ line. The first litter, derived from chimera 1 (90% chimerism) mated with a C57BL/6J wildtype female, was assayed for the targeted deletion of fetuin-B (Figure 3.9). Three of ten siblings showed heterozygosity of the fetuin-B gene, resembling the wildtype and the mutated fetuin-B allele.

Summarizing the results: Three chimeras were generated by injecting manipulated ES cells into blastocysts. Two of these, both containing Knut 1 cells, were germ line transmitting. The chimera containing BAA cells did not transmit these cells to its offspring. Therefore, the establishment of the fetuin-B null-mutant mouse line was restricted to the 129/SvJ x C57BL/6J mixed genetic background of the Knut-1-derived offspring. To develop a mu- tant mouse strain on a pure genetic background the mice were subsequently backcrossed to the defined genetic backgrounds C57BL/6J and DBA/2. 3.1. GENERATING A FETUIN-B-DEFICIENT MOUSE STRAIN 57

4.4 kb 2.3 kb

Figure 3.9: Fetuin-B genotype analysis: tail-DNA taken from the chimera’s first offspring was screened for heterozygous fetuin-B deficiency. Lane 1-10: offspring, thereof 1, 4 and 9 heterozygous fetuin-B genotype; lane 11: positive control (genomic DNA and equimolar masses of the knockout control vector); lane 12: negative control)

3.1.6 Embryo-Transfer and Breeding

In order to keep the generated transgenic animals in a pathogen-free environ- ment, an embryo transfer to the specific pathogen free area (SPF) of the an- imal facility was performed. Heterozygous fetuin-B-deficient offspring of the chimera (N1) were mated to receive homozygous fetuin-B null mutant mice (N1/F2). Five F2 male homozygous fetuin-B knockouts were mated with wildtype C57BL/6J females. The blastocysts were isolated and implanted into SPF foster mothers. By this means, two male pups heterozygous for the fetuin-B deficiency were born in the SPF. These animals resemble the second backcrossing generation (N2) onto the defined genetic background C57BL/6J. From here on, the breeding of the fetuin-B-deficient mice was performed in the SPF facility aiming at generating two distinct fetuin-B-deficient mouse strains with pure genetic backgrounds, C57BL/6 and DBA/2. The C57BL/6 mouse strain is one of the most commonly used laboratory mouse strains often employed as genetically modified models of human diseases. However, DBA/2 mice are prone to calcification and were shown to exhibit a strong calcification phenotype in combination with fetuin-A deficiency. The pedi- grees for C57BL/6 and DBA/2 backcrossing are shown in the Appendix, in Figure 4.2 and 4.1 respectively. 58 CHAPTER 3. RESULTS

3.1.7 Confirmation of Fetuin-B-deficient Mice The deletion of the fetuin-B gene in mice was shown on the genomic level by PCR analysis. The effect of the targeted deletion on the level of protein expression was assayed in blood and tissue using Western blot analysis and immunohistochemistry (Figure 3.10). The fetuin-B protein is absent in the blood of homozygous knockout mice and half the normal amount of fetuin- B protein is present in the blood of heterozygous knockout mice, while the amount of fetuin-A, which was used as reference protein, does not vary (Fig- ure 3.10 A-C).

A +/+ +/+ +/- +/- -/- -/- D WT KO

B

C 120 100

80

% 60

40

20

0 +/+ +/+ +/- +/- -/- -/- Fetuin-A Fetuin-B

Figure 3.10: Fetuin-B knockout analysis on the protein level. Immunodetec- tion of fetuin-B (A) and fetuin-A (B) in sera of wildtype (+/+), heterozygous (+/-) and fetuin-B-deficient (-/-) mice. (C) Densitometric analysis of the amounts of protein detected in the Western blots in A and B. (D) Immunohis- tochemistry of the tongue. Left wildtype (WT), right fetuin-B-deficient (KO) section. Note the bright red staining in the WT section compared to the dark red background staining in the KO section. 3.2. PHENOTYPING FETUIN-B-DEFICIENT MICE 59

Further, no fetuin-B protein can be detected in the tissue of fetuin-B- deficient mice, as shown in Figure 3.10D by the secretory tissue of the tongue known to express high amounts of fetuin-B [15]. Additionally, the translation of partial protein products were undetectable as no protein of lower molecular weight could be detected by the polyclonal anti-fetuin-B antibody (Figure 3.10 A). Thus, the gene knockout was confirmed on the protein expression level of serum proteins proving that the deletion of exon 2 and 3 including the translation start codon indeed effected a true null mutation of fetuin-B.

3.2 Phenotyping Fetuin-B-deficient Mice

After five generations (N5) of heterozygous backcrossing onto the defined ge- netic background C57BL/6J, heterozygous mice (F1-generation) were mated to receive homozygous fetuin-B-deficient, heterozygous fetuin-B deficient and wildtype littermates (F2). As shown in Figure 3.11 the F2 generation con- sisted of 154 individuals, altogether 27 % homozygous fetuin-B-deficient, 25 % wildtype and 48 % heterozygous mice. The genotype distribution was consistent with the expected mendelian heredity ratio of 25:50:25. Therefore, these numbers indicated that fetuin- B-deficient mice were vital and did not suffer of prenatal mortality. The F2 population was used for the following phenotyping procedure.

total 157

40 +/+ 75 +/- 42 -/-

15 m 25 f 36 m 39 f 18 m 24 f

Figure 3.11: Segregation Analysis. Organization chart showing the consis- tence of the F2-generation in numbers. +/+ = wildtype; +/- = heterozygous; -/- = fetuin-B-deficient mice. m = male; f = female. Seven mating cou- ples (F1), 14 heterozygous females and 7 heterozygous males gave rise to the population. 60 CHAPTER 3. RESULTS

3.2.1 Fertility

Evaluating the fertility of fetuin-B-deficient mice had become necessary be- cause the first matings of homozygous fetuin-B-deficient mice failed to pro- duce any litters. Increasing the number of mating couples still did not lead to successful mating, while, at the same time wildtype littermates were fertile as shown in Table 3.2.

Table 3.2: Numbers of wildtype (+/+) and homozygous fetuin-B knockout (-/-) matings, duration and outcome. The mating of 8 female and 7 male homozygous fetuin-B-deficient mice in a total of 13 matings (duration between 1 and 8 months) did not lead to any offspring. However, 6 female and 4 male wildtype littermates gave rise to 88 pups resulting from 6 matings that lasted 3 to 6 months.

Mating ] of ] of mating ] of ] of pups litter female x male matings days litters size FetuB-/- 1 242 - - 0 2 106 - - 0 1 93 - - 0 4 84 - - 0 1 48 - - 0 4 32 - - 0 total: n=8/7 13

FetuB+/+ 2 185 2 14 7 4 106 9 74 8.2 total: n=6/4 6

To explore whether the observed reproductive failure was sex specific, fetuin-B-deficient females were mated with heterozygous and wildtype males and fetuin-B-deficient males were mated with heterozygous and wildtype females. The results of these matings are shown in Table 3.3. 3.2. PHENOTYPING FETUIN-B-DEFICIENT MICE 61

Table 3.3: Outcome of fetuin-B-deficient female and male mice mated with wildtype and heterozygous mice. Female fetuin-B-deficient mice (-/-) did not deliver when mated with neither wildtype (+/+) nor heterozygous (+/-) males. On the contrary, fetuin-B-deficient males (-/-) where able to father offspring when mated to wildtype (+/+) or heterozygous (+/-) females.

Mating ] of ] of ] of ] of pups litter female x male matings plugs pregnancies size -/- (n=9) x +/+ (n=5) 9 9 0 0 0 -/- (n=4) x +/- (n=4) 4 4 0 0 0 +/+ (n=9) x -/- (n=9) 9 9 8 50 6.25 +/- (n=4) x -/- (n=4) 4 4 3 12 4 +/- (n=7) x +/- (n=4) 7 6 4 18 4.2 +/+ (n=4) x +/+ (n=2) 4 4 4 29 7.25

To verify the copulation activity breeding females were routinely checked for copulation plugs in the morning. By this token no differences concerning the mating activity of wildtype and fetuin-B-deficient males were observed. While knockout females mated with wildtype or heterozygous males failed to breed, knockout males readily sired offspring when mated with wildtype or heterozygous fetuin-B-deficient females.

The concatenated protocols and the outcome of the total number of mat- ings of the fetuin-B-deficient mice and their wildtype litter mates are depicted in Figure 3.12.

Summarizing the results: Homozygous fetuin-B-deficient mice copulated but failed to produce offspring. Female mice deficient for fetuin-B were in- fertile regardless of the genotype of the mate, while fetuin-B-deficient males were fertile. Therefore, the breeding defect was due to female infertility. 62 CHAPTER 3. RESULTS 252 252 03

198 198 199 199 229 230 230 171 171

258 255 250 170 165 157 155 154 153

litter 02 7 92 109 120 11 221 229 8 9 7 6

01 78 84

mating 12 7 11 120 109 84 78

11

10 32 56

71 68 22

09 fetuin-B knockout 08

07 20 18 10 7 8 6 9

223

06 heterozygot 241

04 05 166

wildtype

165 03

167 heterozygote females knockout females wildtype females

Figure 3.12: Mating Calendar. Graphical overview showing numbers of matings and litter birth for one year. Each mating is indicated by a black line. Circles symbolize females, circles including an arrow symbolize male individuals. Black closed circles indicate wildtype, black half-closed circles indicate heterozygous and open circles indicate fetuin-B-deficient individuals. Numbers identify each animal. The birth of litters are indicated by stars. This graphical overview demonstrates that fetuin-B-deficient mice did not produce any offspring when the female was homozygous fetuin-B deficient, whereas wildtype and heterozygous fetuin-B-deficient mice delivered offspring during the same time period. 3.2. PHENOTYPING FETUIN-B-DEFICIENT MICE 63

Oocytes

Histological evaluation of knockout uteri and ovaries did not reveal any ab- normalities when compared to wildtype littermates (see Figure 3.18). On both sections advancing stages of growing follicles were visible. Though, it was questioned, if the infertility of fetuin-B-deficient females was due to missing ovulation. For this reason, ovulation of naturally mated wildtype and fetuin-B-deficient females was examined by dissecting the upper part of the oviduct (the am- pulla) at day 0.5 pc. Eight wildtype and ten fetuin-B-deficient females were examined. Hereby, ovulated oocytes were isolated from both genotypes. The average number of isolated oocytes per wildtype and fetuin-B-deficient fe- male was 8 ± 3. Consequently, the female infertility could not be due to an ovulation defect.

Figure 3.13: (A) Frequency distribution of the oocytes’ size including the zona pellucida (n=78) and (B) thickness of the zona pellucida (n=17). Mi- crographs of oocytes were used to measure their diameter and zona pellucida thickness (DISKUS software). 64 CHAPTER 3. RESULTS

Figure 3.14: Micrographs of in Vitro cultivated oocytes isolated from nat- urally mated females. Oocytes isolated from wildtype females at day 0.5 (A,B,E,F), 1.5 (I) and 2.5 pc (K). Oocytes isolated from fetuin-B-deficient females at day 0.5 (C,D,G,H), 1.5 (J) and 2.5 pc (L). 40 x magnification, light microspcopy with differential interference optics. Note that oocytes derived from fetuin-B-deficient females remain in the single-cell stage and do not de- velop into the two-cell stage of an embryo. Additionally, the morphology of these oocytes is altered: the perivitelline compartment is not visible. 3.2. PHENOTYPING FETUIN-B-DEFICIENT MICE 65

The isolated oocytes showed reproducible differences concerning their mor- phology. Oocytes isolated from wildtype females displayed fertilization char- acteristics such as pro-nuclei and a pair of polar bodies (Figure 3.14 A, B, E, F). These characteristics were not visible in oocytes isolated from fetuin-B knockouts. Furthermore, the perivitelline compartment was not visible in these oocytes, due to the zona pellucida that surrounded these oocytes very tightly (Figure 3.14 C, D, G, H). When measuring the diameter of the oocytes, including the zona pellucida, it became obvious that oocytes derived from fetuin-B deficient mice where smaller by 11% compared to oocytes isolated from wildtype females (92.6 µm ± 2.8 µm and 103.5 µm ± 3.8 µm). The size distribution of 78 evaluated oocytes are shown in Figure 3.13 A. The thickness of the zona pellucida (measured at 17 oocytes) was decreased significantly by 26% (p= 0.0003) for oocytes isolated from fetuin-B-deficient females (Figure 3.13 B). However, the diameter of the cytoplasm was 4-8% larger in oocytes derived from fetuin-B deficient females than in oocytes derived from wildtype females. Keeping the isolated oocytes in vitro enabled exploring their developmen- tal potency as shown in Figure 3.15.

Figure 3.15: Kaplan-Meier developmental curve of oocytes. Numbers of oocytes developing into progressive stages of pre-implantation embryos. The Oocytes of wildtype and fetuin-B-deficient females were isolated at 0.5 dpc, further cultivated in vitro and evaluated daily by light microspcopy with dif- ferential interference optics. 66 CHAPTER 3. RESULTS

While 79% (48/61) of oocytes isolated from wildtype females developed into a two-cell stage within 24 h, 98% (78/81) of the oocytes that were isolated from fetuin-B-deficient females remained undivided. Figure 3.14 shows representative micrographs of the in vitro cultivated cells. The three oocytes (2% totally) isolated from fetuin-B-deficient mice that developed to a two-cell stage could further be classified as parthenogenetic. Micrographs of these cells are shown in Figure 3.16. This data indicated, that there was a fertilization defect.

Figure 3.16: Micrographs of parthenogenetically developed oocytes derived from naturally mated fetuin-B-deficient females (E-H, I-L, M+N)) compared to representative micrographs of fertilized oocytes derived from wildtype fe- males (A-D). From left side to right side: day 0.5, 1.5, 3 and 5 pc. . 3.2. PHENOTYPING FETUIN-B-DEFICIENT MICE 67

3.2.2 Anatomical and Morphological Analysis Primary Morphological Analysis

The morphological analysis according to Fuchs et al. [61] yields first-line in- formation about skeletal malformations. Fetuin-B-null mice, resulting from heterozygous matings, showed normal behavior in their cage. Apathetic or hyperactivity disorders were absent. Their locomotion was not altered. Fur- thermore, no difference in size or grooming behavior was identified compared to wildtype litter mates. By observing the mice in more detail, while sit- ting on the experimentator’s hand, the natural habit of mice to explore their environment was noted, always trying to achieve an upright position. Head shape, vertebral column, pelvis elevation and tail movement appeared nat- ural. Holding the animal by its neck the shape of the rib cage, the limbs and also the vertebrae of the tail were free of major malformations. Visual inspection and palpation suggested that limbs, joints, vertebral ribcage, tail mandibles and teeth were all normal and free of gross anatomical malforma- tions.

X-Ray Analysis of Skeletal Morphology

In order to detect putative skeletal abnormalities conventional X-ray pictures were taken and scored for congenital bone diseases, signals of infections as well as joint dislocations. X-ray pictures were taken of 12 and six month old knockouts (n=6). Figure 3.17 shows representative X-ray pictures of 12 months old fetuin-B-deficient and wildtype mice. The skeletal anatomy appeared normal. Furthermore the X-ray pictures revealed that the fetuin- B-deficient mice did not suffer of ectopic calcification disorders like mice deficient in the related fetuin-A protein.

Necropsy and Histology

The post-mortem examination (necropsy) of the fetuin-B-deficient mice was performed to further characterize the clinical features of the genetic modi- fication. In contrast to the primary morphological analysis, necropsy is an invasive method which evaluates the organs within the body. The aim is to identify abnormalities that are due to the genetic modification. The necropsy was performed with matured six-month-old wildtype and 68 CHAPTER 3. RESULTS

Figure 3.17: Representative X-rays of 12 months old wildtype (A) and fetuin-B-deficient mice (B). Note that there are neither signs for skeletal mal- formations, nor calcification disorders, a characteristic of fetuin-A deficient mice. 3.2. PHENOTYPING FETUIN-B-DEFICIENT MICE 69 fetuin-B-deficient males and females. Each group consisted of three individ- uals. The fetuin-B-deficient mice revealed no gross morphological differences when carefully compared to the wildtype litter mates. The size and weight of the organs were inconspicuous. For histological analysis, the organs were fixed, sectioned, hematoxylin-and eosin-stained and evaluated by means of light microscopy. Selected representative microphotographs are shown in Figure 3.18-3.21. Figure 3.18 A and B show sections of the epididymis of wildtype and fetuin-B-deficient male mice, depicting characteristic spermatozoa in the lu- men of the epididymal ducts which are formed by epithelial lining and smooth muscle cells. The sections of the testis in Figure 3.18 C and D show the proceeding differentiation of the spermatogonia in seminiferous tubules sur- rounded by Sertoli and Leydig cells, suggesting that the action of the repro- ductive organs was not altered in fetuin-B-deficient male mice. Figure 3.18 E and F show sections of the uteri of wildtype and fetuin- B-deficient female mice. The uterine horns are composed of the endome- trial epithelium, endometrial glands and endometrial stroma surrounded by a smooth muscle layer. Sections of the ovary (Figure 3.18 G and H) show different stages of folliculogenesis, suggesting that ovulation occurred in both genotypes. Figure 3.19 shows representative sections of the small intestine (A and B), special lymphoid tissue located within the intestine, the Peyer’s patches (C and D) and sections of the large intestine (G and H). Figure 3.20 A and B show representative micrographs of wildtype and fetuin-B-deficient liver sections. Both tissues are characterized by an uneven size of hepatocytes and hepatocellular nuclei. Figure 3.20 D and E show representative sections of the kidney. The functional structures of the kidney, the glomeruli, as well as proximal convoluted tubules are visible and suggest normal function. Figure 3.20 E and F show representative sections of the pancreas and Figure 3.20 G and H of the spleen. Figure 3.21 A and B show sections of the outer ear and of the lung (C and D). The latter micrographs reveal physiological structures such as alveolar ducts and bronchioles. In summary, the fetuin-B-deficient tissue analyzed did not reveal gross morphological abnormalities or pathophysiological conditions. 70 CHAPTER 3. RESULTS

Figure 3.18: Representative micrographs of male and female reproductive organs: epididymis (A, B) and testis (C, D); uterus (E, F) and ovary (G, H) of wildtype (A,C,E,G) and fetuin-B-deficient (B, D, F, H) mice (4µm HE sections, scale bar: 200 µm). 3.2. PHENOTYPING FETUIN-B-DEFICIENT MICE 71

Figure 3.19: Representative micrographs of the intestines of wildtype (A, C, E, G) and fetuin-B-deficient (B, D, F, H) mice: duodenum (A, B), Peyer’s Patch (C, D), jejenum (E, F), colon (G, H) (4µm HE sections). 72 CHAPTER 3. RESULTS

Figure 3.20: Representative micrographs of liver (A, B), kidney (C, D), pancreas (E, F) and spleen (G, H) of wildtype (A, C, E, G) and fetuin-B- deficient (B, D, F, H) mice (4µm HE sections). 3.2. PHENOTYPING FETUIN-B-DEFICIENT MICE 73

Figure 3.21: Representative micrographs of ear (A, B) and lung (C, D) of wildtype and fetuin-B-deficient mice (4µm HE sections). 74 CHAPTER 3. RESULTS

3.2.3 Physiological Parameters General physiological parameters such as body weight, blood pressure and pulse rate were measured to determine the physical condition of the fetuin- B-deficient mice.

Weight: The weight measurement was performed weekly from three weeks up to four months of age. Figure 3.22 shows the plotted data. The analysis of the accumulated data revealed that there was no difference between the weight of sex and age matched wildtype and fetuin-B knockout mice.

Blood pressure and pulse rate: Blood pressure and pulse rate were measured using the ”BP-2000 Blood pressure Analysis System” (Visitech Systems). Sex matched wildtype and fetuin-B-deficient mice, three months of age were analyzed. Each group consisted of seven animals. After a training phase, all measurements that yielded data for systolic blood pressure (SBP), diastolic blood pressure (DBP) and pulse rate were plotted. Data series with individual measurements missing were discarded entirely. The resulting data are depicted in Figure 3.23. The median blood pressure value of wildtype and fetuin-B-deficient males was 97/71 (SBP/DBP). The median of female mice was 100/73 for wildtype and 100/75 for fetuin-B-deficient females. The median pulse of wildtype males (513 bpm) and fetuin-B-deficient males (499 bpm) were inconspicuous. The median pulse of the female mice was slightly faster (577 bpm for wildtype and 600 bpm for fetuin-B-deficient females). Taken together the pulse rate and blood pressure measurements indicated that the fetuin-B-deficient mice have no gross cardiovascular defects affecting these parameters.

3.2.4 Glucose Tolerance Test A glucose tolerance test was performed because an initial serum analysis for glucose levels had given a hint for impaired glucose metabolism in male fetuin- B-deficient mice. Another reason for analyzing the glucose metabolism in fetuin-B-deficient mice was that fetuin-A has been shown to mediate insulin receptor signaling [67, 68, 69] and fetuin-A-deficient mice showed an improved insulin sensitivity when compared to wildtype mice [28]. 3.2. PHENOTYPING FETUIN-B-DEFICIENT MICE 75

Figure 3.22: Body weight of sex matched wildtype and fetuin-B-deficient mice. Each dot represents the mean value of weight measurements at that day. The curve was interpolated using a non-linear regression fit of Graph Pad Prism. 76 CHAPTER 3. RESULTS

Figure 3.23: Measurement of blood pressure and pulse rate. Every dot represents one measurement value of age and sex matched wildtype and fetuin- B-deficient mice (n=7 for each group: male +/+, -/-, female +/+ and -/-). The median is depicted by a black line. 3.2. PHENOTYPING FETUIN-B-DEFICIENT MICE 77

Figure 3.24 depicts the glucose metabolism of age and sex matched wild- type and fetuin-B knockout mice after a challenge of 2 g/kg body weight of 20 % D-glucose. A

B

Figure 3.24: Glucose tolerance test. Comparison of glucose metabolism of wildtype (WT) and fetuin-B-deficient (KO) males (A) and females (B). n=10 for each group. Note that there were no statistically significant differences between the genotypes at any timepoint.

The resulting data were analyzed using an impaired two-sided t-test and Graph Pad Prism software. The analysis revealed that the wildtype and fetuin-B-deficient mice did not differ significantly in their glucose metabolism (P=0.8 and 0.7 comparing wildtype and fetuin-B-deficient females and wild- type and fetuin-B-deficient males, respectively). Thus, fetuin-B deficiency was not associated with a diabetic phenotype of hyperglycemia or glucose intolerance. 78 CHAPTER 3. RESULTS

3.2.5 Clinical Chemical Screening

The clinical serum chemistry screening is a diagnostic tool suitable for de- tecting defects in organ systems, haematological disorders and changes in metabolic pathways. Moreover, the loss of a serum protein could result in impaired major serum functions such as for instance the coagulation system, the osmotic pressure or homeostasis. Therefore, the primary clinical chem- istry screen was chosen such that alterations in a broad range of conditions could be detected with a reliable number of parameters.

Serum analysis: 19 parameters were analyzed from age matched wildtype (n=6) and fetuin-B-deficient (n=6) sera. These parameters included indica- tors for liver, renal, pancreas, and thyroid functions, as well as muscle, bone, cardiovascular system and metabolic disorders. The resulting data are shown in Figure 3.25. The data were analyzed using a Student’s t-test. When comparing wildtype to fetuin-B-deficient sera, none of the 20 parameters reached statistical significant differences.

Coagulation analysis: fetuin-B-deficient plasma was screened for coagu- lation defects. Global tests were chosen that give first line information about the function of the intrinsic coagulation system. The fetuin-B-deficient sera were compared to sera of age and sex matched wildtype litter mates (six individuals each, consisting of three males and three females). By comparing the thromboplastin time (Quick), partial thromboplastin time (PTT), and levels of Fibrinogen and anti-thrombin III no conspicuous differences were detected in the function of the intrinsic coagulation system of wildtype and fetuin-B-deficient mice. The thromboplastin time and the level of fibrinogen were slightly elevated in male knockouts. Figure 3.26 shows the data in a graphical overview. In summary, the serum chemistry did not reveal any major alterations in organ function or blood clotting associated with the lack of fetuin-B. The data suggests that fetuin-B-deficient mice are basically healthy without any severe clinical disorders. 3.2. PHENOTYPING FETUIN-B-DEFICIENT MICE 79

Figure 3.25: Clinical chemistry screening. Comparison of wildtype (black bars) and fetuin-B-deficient (white bars) sera on a logarithmic axis (n=6 per group). (A) Serum electrolytes: phosphate (PO4) , calcium (CA) , potas- sium (K) and sodium (NA). (B) Enzymes as markers for liver and pancre- atic function: glutamate oxalacetate transaminase (GOT), glutamate pyru- vate transaminase (GPT), gamma glutamyl transferase (GGT), alkaline phos- phatase (AP), amylase(AML), and lipase (LIP). (C) Substrates and total pro- tein: glucose (GLUC), creatinine (CREA), urea (UREA) , cholesterol (CHOL), triglycerides (TRIG), uric acid (HS), bilirubin (BILI) and total protein (TP). Statistical analysis performed using two-sided Students t-test and Microsoft Excel software. 80 CHAPTER 3. RESULTS

Figure 3.26: Blood Coagulation Parameter. Dots represent single measure- ments. Lines represent the median values. Note that thromboplastin time was different in male and female mice, however, this is consistent for wildtype and fetuin-B-deficient mice.

3.2.6 Summarizing the Phenotyping Results

The initial characterization of the fetuin-B-deficient mice revealed female infertility while the reproductivity of fetuin-B deficient males was not altered. It was shown that oocytes derived from fetuin-B-deficient females did not develop into two-cell embryos.

Differences in the anatomy and morphology of these mice were not found when compared to wildtype mice. Furthermore, the fetuin-B deficient mice showed typical physiological parameters when assayed for body weight, blood pressure and pulse rate. The glucose metabolism was not affected by the fetuin-B null mutation. Lastly, the serum analysis covering important func- tional parameters of major organs suggests that fetuin-B-deficient mice are basically healthy without any severe clinical disorders. 3.3. RECOMBINANT MURINE FETUIN-B 81

3.3 Recombinant Murine Fetuin-B

Up to now murine fetuin-B was not available as a purified protein but there was a need for the protein in order to perform functional in vitro analysis. The aim of the following work was to produce an adenovirus that could efficiently transfect the fetuin-B cDNA into mammalian cells for subsequent purification and analysis of the recombinant protein.

3.3.1 Cloning Recombinant Murine Fetuin-B

The final goal was to insert the murine fetuin-B cDNA into an adenoviral vector system. But first, the fetuin-B cDNA had to be equipped with a constitutively active promoter and a purification tag because the adenoviral vector system did not contain a promoter and because the fetuin-B protein was supposed to become an easily purified fusion protein. Therefore, the fetuin-B cDNA was inserted into the eukaryotic expression vector pcDNA3.1- V5/His A (Invitrogen). This vector offers a strong constitutive promoter (CMV) and it provides a c-terminal purification tag (His) that is followed by a poly-adenylation signal. The cloning strategy and the illustration of the employed vectors are shown in Figure 3.27 and are explained in the following: The mouse fetuin-B cDNA clone MGC:25848 (BC018341), which includes the complete FetuB coding sequence, was purchased from Life Technologies, Inc. The Fetuin-B cDNA sequence was amplified by PCR. The primers that were used were supplied with unique restriction sites: the sense primer contained an EcoRI restriction site and was designed to bind upstream from the translation-start codon, and the anti-sense primer contained an AgeI restriction site. This primer was designed to bind to the last codons of the cDNA but excluding the internal stop codon. Figure 3.28 shows the fetuin-B cDNA sequence and the primer-binding sites. Subsequently, the PCR product was ligated to the pGEMT-Vector (Promega). This vector does not have restriction sites for EcoRI and AgeI. Consequently, by digesting the construct with these restriction enzymes, the cloned fetuin-B cDNA sequence, beginning from the translation start codon but missing the internal stop codon, was excised. Finally, the modified fetuin- B cDNA was ligated to the EcoRI/AgeI linearized expression vector. By 82 CHAPTER 3. RESULTS

fetuin-B

EcoR Age I fetuin-B I

DraIII pGEM-T MfeI

CMV fetuin-B His-Tag

EcoRV

p∆E1sp1A adenoviraladenoviral Shuttle Vector backbonebackbone vector vector

recombinant Ad5-CMV- fetuin-B

Figure 3.27: Sequence of cloning the fetuin-B fusion protein beginning with the cloning of fetuin-B cDNA into the PCR product cloning vector pGEMT, then, into the eukaryotic expression vector pcDNA3.1/V5-His in-frame with the histidine purification tag. Finally, the fetuin-B cDNA including CMV promoter and His-tag was cloned into the adenoviral shuttle vector which was recombined with the adenoviral backbone vector to become a recombinant fetuin-B containing Ad5 adenovirus. 3.3. RECOMBINANT MURINE FETUIN-B 83

EcoRI AGAATTCTTACAGTG

AgeI ACCGGTCAT

Figure 3.28: Fetuin-B cDNA amplification primers and their binding sites on the fetuin-B cDNA. The nucleotide sequence upstream +1 does not align to the fetuin-B reference sequence (NM 021564), therefore, the primer was changed accordingly. Note that the UGA stop codon was replaced by a short linker and a His-tag to facilitate recombinant protein purification.

digesting the vector with EcoRI/AgeI the His-tag was connected as close as two additional base triplets with the 3’-end of the fetuin-B cDNA sequence.

The complete coding sequence including the transition from the coding sequence to the vector was sequenced, starting upstream and downstream from the fetuin-B coding sequence within the vector sequence. This showed firstly, that the fetuin-B coding sequence was not mutated, due to the PCR amplification and secondly that the transition from the coding sequence to the vector sequence coding for the His-Tag was in frame as shown in Figure 3.29.

The construct was assayed for expression of fetuin-B by transfecting COS-7 cells and analyzing the cell culture supernatant 36h after transfection with means of Western blotting. While there was no fetuin-B detectable in the mock control (pcDNA3.1-V5/His A) fetuin-B was detected in the media of pcDNA3.1-CMV-FetB-His transfected cells as shown in Figure 3.30.

After having proven the construct´s functionality the complete functional unit consisting of the CMV promoter, the fetuin-B cDNA, the connected His-tag and a polyadenylation signal was excised by the restriction enzymes MfeI and DraIII (Figure 3.27). This fragment was then ligated to the EcoRV opened shuttle vector, p∆E1sp1A. 84 CHAPTER 3. RESULTS

consens rec native

consens rec native

consens rec native consens rec native consens rec native consens rec native consens rec native consens rec native consens rec native consens rec native consens rec native consens rec native 3.3. RECOMBINANT MURINE FETUIN-B 85

consens rec native consens rec native consens rec native consens rec native consens rec native consens rec native consens rec native consens rec native consens rec

Figure 3.29: Consensus sequence of sequencing data (consens), and align- ment of the resulting amino acid translation (rec) and the protein sequence (native). The vector pcDNA3.1-CMV-FetB-His was sequenced using T7 and BGH primers as sense and anti-sense primers, respectively. The sequenced cDNA sequence was not mutated by the PCR amplification; furthermore, the endogenous stop codon was excluded and the His purification tag was con- nected in-frame with the Fetub open reading frame. The putative N-terminus of the secreted protein is marked by the black linings. 86 CHAPTER 3. RESULTS

Figure 3.30: Fetuin-B Western blot of transfected COS-7 cells. Supernatant (10 µl) of pcDNA3.1-CMV-mFetB-His (1) and pcDNA3.1-V5/His A (2) trans- fected COS-7 cells. Murine serum (1:100) was applied as a positive control (3). Right: molecular weight marker

3.3.2 Generating a Fetuin-B Transfecting Adenovirus

The recombinant shuttle vector and the adenoviral backbone vector (shown in Figure 3.27) were homologously recombined by co-transfecting them into 293 cells. These cells produce E1 proteins, which are not encoded by the adenoviral vector. Hence, only these cells enable the assembly of the aden- ovirus.The virus containing supernatant was transferred to COS-7 cells. The proper recombination of shuttle and backbone vector was assayed by detect- ing the recombinant fetuin-B protein in the cell culture supernatant.

3.3.3 Expressing and Purifying Recombinant Fetuin-B

In order to purify the recombinant fetuin-B fusion protein, 45 x 10cm-dishes containing COS-7 cells were treated with the fetuin-B transfecting aden- ovirus. By this, 450 ml (serum-free) cell culture supernatant were harvested, containing the fusion protein. Using the AKTAdesign¨ liquid chromatogra- phy system and HisTrap affinity columns from GE Healthcare, the fetuin-B fusion protein was purified from the supernatant. Half of the supernatant (225 ml) were pumped over the 1 ml HisTrap column (first run), then the column was washed and the fusion protein was eluted. The second half of the supernatant was proceeded accordingly (second run). The flow-through was pooled and pumped over two connected 1 ml HisTrap colums (third run), otherwise it was proceeded as before. Subsequently, the flow-through of the 3.3. RECOMBINANT MURINE FETUIN-B 87 third run was proceeded as the former run. A representative chromatogram of a purification run is shown in Figure 3.31.

Figure 3.31: Representative purification chromatogram. Red line: absorp- tion; black line: conductivity. The absorption plateau resembles the flow through of the cell culture supernatant, the following peak resembles the elu- tion of proteins that had specifically bound to the HisTrap column.

Supernatant, flow-through and the pooled fractions of the purified fetuin- B fusion protein were analyzed by Coomassie staining of a polyacrylamide gel and by a fetuin-B Western blot, both shown in Figure 3.32A and B, respectively. Coomassie staining and Western blot illustrated the purification of the fetuin-B fusion protein: firstly, the quantity of the flow-through band 1 to 4 stays the same in the Coomassie stain, while the amount of fetuin-B decreases with every purification step as shown by the Western blot and secondly, the Coomassie stain shows only one band in the pooled purification fraction, that is not visible in the cell-culture supernatant, identified as purified fetuin-B by the Western blot. The Western blot demonstrates, progressive depletion of fetuin-B from the cell-culture supernatant. After the fourth run fetuin-B was no more detectable in the flow-through. 88 CHAPTER 3. RESULTS

A

1 2 3 4 5 6 7 8 kD

68

43

B 1 2 3 4 5 7 kD

68

43

Figure 3.32: Analysis of purification products. (A) Coomassie stained poly- acrylamide gel (25 µl sample per lane) and (B) anti-fetuin-B Western blot (10 µl sample per lane) of: 1) cell culture supernatant; 2) HisTrap flow-through of the first run; 3) HisTrap flow-through of the third run; 4) HisTrap flow- through of the fourth run; 5) pooled protein containing fractions; 6) pooled protein containing fractions after desalting; 7) murine serum control; 8) fetuin- B deficient serum.

The pooled fetuin-B fractions were dialyzed in PBS overnight. The con- centration of the purified fetuin-B was determined at 0.32 mg/ml by OD280 measurement in comparison to a bovine fetuin-A standard. 3.3. RECOMBINANT MURINE FETUIN-B 89

Sequencing the Recombinant Fetuin-B Protein

Secreted proteins have a short N-terminal sequence, the leader sequence or , that mediates the passage through the cell membrane. During the passage through the ER-Golgi compartment the signal peptide is cleaved by an enzyme, the signal peptidase, which implies that the secreted protein begins with a different N-terminus. By computer analysis a potential signal peptide was identified at the N- terminus of the primary fetuin-B amino acid (AS) sequence. The signal peptide was predicted to encompass residues 1-18 according to the ExPASy database, UniProtKB/Swiss-Prot entry Q9QXC1 (http://au.expasy.org/ /Q9QXC1). To identify the N-terminus of the secreted fetuin-B pro- tein the purified fetuin-B fusion protein was sequenced. The N-terminal se- quence was determined as arginine (Arg), serine (Ser), proline (Pro), proline (Pro) and alanine (Ala). This result experimentally confirmed the predicted signal peptide of the first 18 AS. The complete AS sequence is shown in Figure 3.29. In addition peptide mass fingerprints derived from mass spectroscopy of the purified protein unequivocally proved the identity of the recombinant protein as mouse fetuin-B. 90 CHAPTER 3. RESULTS

3.3.4 Precipitation Inhibition Assay Fetuin-A is a potent inhibitor of calcification. The calcium phosphate pre- cipitation inhibition assay is a well established in vitro method to study the effect of fetuin-A on precipitation of calcium phosphate from a saturated calcium phosphate solution [63, 11]. Since fetuin-B is the closest relative of fetuin-A, it was analyzed if fetuin-B could also inhibit the precipitation of calcium phosphate in vitro. The first assay was performed by measuring the amount of calcium and phosphate precipitate in the presence of 1-5 % fetuin- B deficient serum, comparing its effect to wildtype and fetuin-A-deficient serum and a protein-free solution as negative control. Figure3.33 A shows that under the conditions of the assay wildtype and fetuin-B-deficient serum effected full precipitation when admixed at 1% to the precipitation mixture. In contrast fetuin-A deficient serum had to be admixed at a minimum of 3% [vol/vol] to effect a similar inhibition. Thus fetuin-B unlike fetuin-A is not an efficient inhibitor of calcium phosphate precipitation. Purified recombinant mouse fetuin-B likewise failed to inhibit the precipitation of calcium phos- phate (Figure 3.33 B), confirming the result obtained with fetuin-deficient sera and with recombinant fetuin-B expressed in bacteria [11, 15].

3.3.5 TGF-β Reporter Activity in Mink Lung Cells Fetuin-A has been shown to modulate TGF-β activity by acting as a soluble TGF-β type II receptor mimic [12]. Fetuin-B revealed sequence homology with the TGF-β type II receptor as described in subsection 1.1.1 (Figure 1.2). Therefore, the interaction of fetuin-B and TGF-β was analyzed in a biological system: the mink lung cell reporter assay. The mink lung cell reporter assay is based on a stably transfected reporter construct that consists of a PAI-1 promoter and a luciferase gene in mink lung epithelial cells (MLEC). The PAI-1 gene is directly activated by TGF-β signaling, therefore, the presence of the PAI-1 promoter makes the mink lung cells TGF-β responsive, with a readout of luciferase activity. Additionally, the expression of luciferase depends on the concentration of TGF-β: the more TGF-β, the more luciferase activity is measurable (Figure 3.34A)[64]. 3.3. RECOMBINANT MURINE FETUIN-B 91

Figure 3.33: Calcium phosphate precipitation inhibition assay. (A) The as- say was performed using wildtype, fetuin-A-deficient (fetuin-A -/-) and fetuin- B-deficient (fetuin-B -/-) sera. Wildtype and fetuin-B-deficient sera inhibited precipitation at low serum concentrations. Fetuin-A-deficient sera show a re- duced inhibition capacity. (B) The assay was performed using bovine fetuin-A and recombinant murine fetuin-B expressed in COS-7 cells. In both cases 100 % precipitation was determined by the protein-free control. Pure fetuin-A inhibited precipitation of calcium phosphate, whereas, pure fetuin-B did not show any inhibitory potential. 92 CHAPTER 3. RESULTS

Figure 3.34: Luciferase activity of mink lung (MLEC) reporter cells. (A) Increasing amounts of TGF-β led to increased luciferase expression; (B) in- creasing amounts of bovine fetuin-A did not induce luciferase expression in the absence of TGF-β while increasing amounts of fetuin-B did induce luciferase expression in the absence of TGF-β. The addition of 250 pg/ml TGF-β to the increasing amounts of fetuin-A and fetuin-B caused luciferase expression in an additive manner. LCPS: luciferase counts per sec. 3.3. RECOMBINANT MURINE FETUIN-B 93

The assay was performed employing a fixed TGF-β concentration of 250 pg/ml. Recombinant murine fetuin-B fusion protein and bovine fetuin-A were added in increasing concentrations. Figure 3.34 B shows the impact of fetuin-B and fetuin-A on TGF-β signaling. Fetuin-B induced luciferase activity in the absence of TGF-β in a concen- tration dependent manner. whereas, fetuin-A did not induce any luciferase activity. In the presence of 250 pg/ml TGF-β again luciferase activity was further increased with increasing concentrations of fetuin-B. In contrast, the baseline luciferase activity of 250 pg/ml TGF-β was not further increased by adding increased amounts of fetuin-A. Thus, fetuin-B induced the luciferase expression, controlled by the PAI- 1 promoter but did not inhibit TGF-β activity. The inhibitory activity of fetuin-A, which was previously shown in this assay for recombinant human fetuin-A, could not be reproduced with bovine fetuin-A for unknown reasons.

3.3.6 Serum Concentration of Fetuin-B Mouse fetuin-B serum levels had been previously estimated employing a prokaryotically expressed MBP-fetuin-B fusion protein [15]. Here, these mea- surements were repeated employing the eukaryotically expressed fetuin-B protein as a standard. Quantitative fetuin-B Western blots were performed in order to estimate the fetuin-B level in mouse serum. Nine adult females and ten adult male mice were analyzed. As shown in Figure 3.35 the average serum concentration of fetuin-B in adult mice was higher in males (406 µg/ml ± 41 ) than females (334 µg/ml ± 93). This is in contrast to the fetuin-B serum levels reported previously in human serum [15] where females had 50% higher serum levels than males. However, in this analysis females showed a broader fetuin-B concentration range, indicating that the concentration may cycle. This needs further investigation. 94 CHAPTER 3. RESULTS

Figure 3.35: Fetuin-B serum concentration in adult mice. (A) Represen- tative Western blot of adult mouse serum (6-10) and recombinant-fetuin-B dilution series (1-5) that were quantified by the ImageJ software in (B) The median fetuin-B serum concentration in males was 0.4 mg/ml ± 41 (n=11). The median serum concentration in females was 0.3 mg/ml ± 93 (n=9).

3.3.7 Summarizing the Results of the in Vitro Data

The fetuin-B cDNA controlled by a constitutively active CMV promoter and elongated by a histidine purification tag was successfully cloned into an adenoviral vector system. The recombinant fetuin-B fusion protein was purified from the cell culture supernatant of transfected COS-7 cells. The N- terminus of the mature fusion protein was sequenced. Thereby, the predicted signal peptide was confirmed experimentally. Employing the purified recombinant fetuin-B protein as a standard the concentration of fetuin-B in mouse serum was assessed by Western blot anal- 3.4. PROMOTER ANALYSIS 95 ysis. The average serum concentration was determined at 0.4 mg/ml for males and 0.3 mg/ml for females. The purified fetuin-B protein did not reveal any calcium phosphate precipitation inhibitory potential unlike it’s closest relative fetuin-A, which is known as a strong inhibitor of calcium phosphate precipitation. Unlike fetuin-A, fetuin-B activated luciferase expression in the mink lung cell reporter assay in the absence of TGF-β. Still further experiments need to elucidate whether the luciferase expression was due to fetuin-B mediated signaling via the TGF-β pathway or whether other mediators were contained in the fetuin-B fraction as a viral contaminant.

3.4 Promoter Analysis

3.4.1 Cloning and Analysis of the Fetub Promoter The promoter controlling Fetub expression has not been studied yet. Com- puter analysis predicted two possible promoter regions for the murine Fetub gene as shown in Figure 3.36 and one putative promoter region was found for human and rat gene.

Figure 3.36: Localization of the two predicted FetuB Promoters. The FetuB gene’s exon/intron structure and the localization of the predicted promoter regions are illustrated in this figure. Each promoter region (red box) is located upstream from each alternative 5’-UTR (for review see Section 1.1.3). MP1: potential mouse promoter #1; MP2: potential mouse promoter #2.

To construct a Fetub reporter assay the two predicted murine promoter regions were amplified by PCR and cloned into a pEGFP-1 vector (BD Bio- sciences Clonetech) as shown in Figure 3.37. 96 CHAPTER 3. RESULTS

KpnI XmaI 600 bp MP1 or MP2

Figure 3.37: Murine fetuin-B promoter constructs. 600 bp of the predicted mouse promoter #1 and #2 were cloned into the promoter less vector pEGFP- 1. The pEGFP-1 vector contains the enhanced green fluorescence protein (EGFP) gene that is only expressed in the presence of an active promoter. The promoter activity can be assayed by transfecting the recombinant vector into target cells and screen for green fluorescing cells.

The reporter constructs were transfected into various cell types (HeLa, 3T3, HaCaT, Hepa1-6 and HepG2), to study Fetub expression in varying cellular contexts. The putative promoter sequence MP1 that is located more distal to the Fetub gene led to the expression of EGFP in all tested cell types, whereas, the putative promoter sequence MP2 that is located more proximal to the Fetub gene did not lead to EGFP expression, when compared to the negative control. This indicates that MP2 is either not functional as a promoter, or that MP2 functions tissue specific, e.g. for the ovary. 3.4. PROMOTER ANALYSIS 97

Figure 3.38: Murine fetuin-B promoter analysis. Two putative fetuin-B promoter regions (MP1 and MP2) were cloned into a EGFP-reporter vector and transfected into the murine and human cell lines indicated on top of the figure. A CMV promoter was used as a constitutive active promoter. The negative control (NC) was a EGFP-reporter vector without a promoter. HeLa: human cervix carcinoma cell line; 3T3: murine fibroblast cell line; HaCaT: human epithelial keratinocyte cell line; Hepa1-6: murine hepatoma cells; HepG2: human hepatoma cells.

Chapter 4

Discussion

4.1 Generation of Fetuin-B-deficient Mice

4.1.1 Gene Deletion

The fetuin-B-gene knockout was achieved by deleting exon 2 and 3 and in- serting a neomycin resistance marker of E. coli origin (neo-cassette). The overall genomic sequence of the fetuin-B gene was thus shortened by 235 bases. The reason for deleting exon 2 and 3 was that two alternative transla- tion start codons were described for the Fetub gene located in these exons. By deleting these exons the translation start codons, required for protein synthesis were removed, regardless of the putative splice variants of primary transcripts. The deletion of exon 2 and 3 was sufficient to prevent the translation of fetuin-B. This was proven on the protein level: fetuin-B was absent in the serum and tissue of fetuin-B-mutant mice.

4.1.2 Embryonic Stem Cells and Genetic Background

The injection of ES cells directly into the cavities of blastocysts is a straight- forward method to generate chimeric mice carrying alleles with a mutant genome [70]. To derive a mutant mouse strain from these chimeric mice, germ-line transmission has to be achieved. This is known to depend heav- ily on the genetic background of the manipulated ES cells and the recipient blastocyst. For example, Balb/c blastocysts have been shown to work well in

99 100 CHAPTER 4. DISCUSSION combination with C57BL/6-derived ES cells [71], and the use of F1 hybrid ES cells have demonstrated a superior developmental potential [72]. However, the advantage of the superior developmental potential of F1 hybrid stem cells is diminished by the disadvantage of obtaining a mutant mouse strain with a mixed genetic background. As a consequence a series of backcrossings with an inbred strain is required to ensure a uniform genetic background. Accord- ingly, in this work F1 hybrid ES cells (129/Sv x C57BL/6) and inbred ES cells (C57BL/6) were targeted and subsequently injected into Balb/c-derived blastocysts in parallel. Their transfection and homologous-recombination rate were identical. However, as described above, the F1 hybrid stem cell was superior to the inbred ES cell because it transmitted the germ-line. As a result, the targeted mutation was obtained on a mixed genetic background. This required time-consuming backcrossing with the inbred strain C57BL/6. At least 10 generations of backcrossing, three months per generation, are re- quired to obtain a uniform genetic background consisting of 99.9% identical genetic loci. The characterization of the fetuin-B-deficient mouse strain was begun on F2 littermates after the fifth backcrossing generation with the C57BL/6 in- bred strain. Statistically 98% of the donor genome was eliminated by that time. Thus, wildtype littermates and not commercial pure-bred wildtype animals were used as control animals. Some of the differences, such as the clinical chemistry values, for example, could thus be due to the remaining genetic divergence among the individuals.

4.1.3 Screening Procedure

After the transfection of the ES cells it was necessary to identify the homol- ogously recombined ES-cell clones. For the first screening, a PCR approach was chosen because it facilitates fast and high-throughput screening com- pared to Southern bloting. However, the PCR absolutely requires a positive control. To this end a positive control was cloned: the knockout control vector. It resembled the gene-targeting vector but consisted of an extended 3’-homologous sequence, thereby mimicking the recombinant fetuin-B allele. The knockout control vector enabled simulation of the PCR screening proce- dure, thus improving performance and enshuring the reliability of the screen- ing system. 4.1. GENERATION OF FETUIN-B-DEFICIENT MICE 101

Southern blot analyses were performed to confirm the PCR-positive clones, again facilitated by the use of the knockout control vector as a positive con- trol. Both analytic procedures were optimized before the targeted ES cell DNA was analyzed. Therefore, it can be safely assumed that all positive ES-cell clones were detected by the screening procedure.

4.1.4 Expression of Fetuin Family Proteins The fetuin-B-null mutation was confirmed on the level of protein expression using Western blot analysis of serum. The absence of the fetuin-B protein was determined in the serum of homozygous fetuin-B-deficient mice and half the normal amount of fetuin-B protein was determined in the blood of heterozy- gous mice. This finding suggests that both Fetub alleles are independently expressed under physiological conditions. It is known that the fetuin-B protein is less abundant in fetuin-A-deficient mice [15]. This was determined not only on the protein level, but also on the level of mRNA expression (unpublished data). The down-regulation of fetuin-B in fetuin-A-deficient mice might be caused by regulatory sequences that were compromised in the course of targeting the fetuin-A gene, since both genes are located in close proximity. Vice versa fetuin-A expression may be compromised in fetuin-B-knockout mice. Thus, the level of fetuin-A protein expression was analyzed in the serum of fetuin-B-deficient mice. The expression of fetuin-A was unchanged in both heterozygous and homozygous fetuin-B-deficient mice. This suggests that the expression of the fetuin-A gene was not compromised by targeting the fetuin-B gene and that the ex- pression of fetuin-A does not depend on the availability of fetuin-B.

4.1.5 Fetuin Serum Concentrations The serum concentration of fetuin-A in adult mice was measured at about 200 µg/ml [13]. Denecke et al. first quantified fetuin-B within mouse serum [15]. They used prokaryotically expressed MGP-fetuin-B fusion protein as a reference protein and determined 156 ± 3 µg/ml fetuin-B within the serum of male mice. Furthermore, they detected that fetuin-B levels are two-fold higher in human females than in males. In this study, eukaryotically expressed instead of bacterially expressed fetuin-B was employed as a standard. Adult male and female mice were 102 CHAPTER 4. DISCUSSION analyzed. The overall fetuin-B concentration measured in this study was higher than determined by Denecke et al. (156 µg/ml compared to 406 µg/ml for males and 334 µg/ml for females) and also higher than the known concentration for fetuin-A. However, in mice the concentration of fetuin-A is known to be highest after birth [25]. This study showed that females show a wider range of serum concentration of fetuin-B when compared to males, implying that fetuin-B could undergo a cyclic regulation in females, possibly following the estrus. To this end further studies need to evaluate whether there is a link between the estrus phase and the serum concentration of fetuin-B.

4.2 Phenotyping

4.2.1 Vitality

The segregation analysis showed that 25% of the F2 offspring were wildtype, 48% were heterozygous and 28% were fetuin-B deficient. This data conforms to Mendel’s second law, predicting 25% wildtype, 50% heterozygous and 25% fetuin-B deficient mice. Firstly, this data showed that fetuin-B-deficient mice are vital. Secondly, they indicated that fetuin-B-deficient mice develop without extraordinary embryonal lethality, since fetuin-B-deficient mice are born at the same rate as their wildtype littermates. Hence, embryo development is independent of embryonic expression of fetuin-B. However, fetuin-B may still be required for proper embryo development since it may be provided by the mother through the placenta. A hint in that direction stemmed from the observation that fetuin-B-deficient females were not able to conceive. Embryo development did not even begin in these mice, as detailed in the following section.

4.2.2 Female Infertility

This work revealed that fetuin-B-deficient females were infertile while het- erozygous females were fertile. Fetuin-B-deficient females did not deliver when mated with any genotype. Copulation activity was normal as vaginal plugs were routinely observed after mating. By the same token, no differ- ences between the copulation activity of wildtype and fetuin-B deficient male 4.2. PHENOTYPING 103 mice were observed. Furthermore, it was shown that fetuin-B-deficient males were able to sire offspring when mated to wildtype and heterozygous females. In conclusion, it was shown that fetuin-B was necessary for female but not for male reproduction. The fact that the targeted deletion of a serum protein that is predomi- nantly expressed by the liver resulted in infertility seems surprising. However, proteins of the extracellular coat surrounding non-cyprinoid fish oocytes are also synthesized by the liver. They are transported by the bloodstream to the ovary where they are incorporated into the vitelline envelope of the growing oocyte. The proteins involved in the vitelline envelope and the mouse zona pellucida are closely related to each other, sharing 25% sequence identity between trout and mouse [73]. Also avian eggs consist of some proteins that are partly synthesized by the liver and others in the ovary [74]. It would appear that during evolution the sites of synthesis included the liver and the ovary. Additionally, the targeted deletion of the serum protein urinary trypsin inhibitor/bikunin that is expressed by the liver resulted in impaired fe- male fertility. This was due to reduced numbers of ovulated oocytes and an unstable cumulus oocyte complex, COC [75, 76]. However, bikunin is the only proteinase inhibitor that has been shown to contribute to female fertility so far according to the mouse genome informatics database, MGI (www.informatics.jax.org). In fact, the MGI database lists 216 targeted genes that contribute to fe- male infertility. These mouse models reveal defects in germ cell maintenance, proliferation and development as in Dazla-deficient mice [77], the formation of primordial follicles as in Figα-deficient mice [78], the formation of secondary follicles as in Gdf9-deficient mice [79], the formation of antral follicles as in FSHβ-deficient mice [80], ovulation problems such as in cyclooxygenase 2 [81] and progesterone receptor deficiencies [82], fertilization problems such as in CD9-deficient mice [52, 50], oocyte-to-embryo transition deficiencies such as in Zar1-deficient mice [36] or implantation failures as in Lif deficient mice [83]. Fetuin-B-deficient females were able to ovulate and histological sections of the ovaries revealed different stages of folliculogenesis as expected for fertile mice. This implies that signaling mediated by the follicle-stimulating hor- 104 CHAPTER 4. DISCUSSION mone and by the luteinizing hormone in the ovarian follicle was not affected in the fetuin-B-deficient mice. The early folliculogenesis was not influenced by the absence of fetuin- B, as questioned in a recent study on newborn mouse ovaries: Choi et al. showed that fetuin-B was expressed during early oogenesis. Interestingly, its expression was significantly down-regulated in ovaries that lacked Nobox, a transcription factor for oocyte-specific genes. Nobox-knockout ovaries are grossly normal at birth, however, the lack of Nobox inhibits oocyte and fol- licle growth beyond the primordial follicle stage [84]. The fetuin-B-deficient mouse model showed that even though fetuin-B was down-regulated in the Nobox deficient oocytes it did not contribute to the observed phenotype be- cause the early folliculogenesis is not altered in the fetuin-B-deficient mice. However, the down-regulation of fetuin-B in the absence of Nobox suggests that Nobox might regulate the oocyte-specific expression of fetuin-B possibly by activating the promoter MP2 (characterized in Section 3.4.1). The ovulated oocytes of fetuin-B-deficient females were well-organized in cumulus-oocyte complexes (COC). These complexes have been shown to be essential not only for the development of the oocyte but also for the fertil- ization potential of the oocytes. However, the ovulated oocytes of naturally mated fetuin-B-deficient fe- males did not develop to the two-cell stage. Morphologically, these oocytes did not reveal any of the characteristics of fertilized oocytes such as two polar bodies and pro-nuclei, furthermore, their zona-pellucida thickness was reduced and a perivitelline space was never observed. The morphological phenotype described above implies that the infertil- ity of fetuin-B-deficient females was most likely due to a fertilization defect and not to a blocked oocyte-to-embryo transition as in mice deficient in the Zar1 (Zygote arrest1, maternal-effect gene) and Zfp36I2 (zinc-finger protein involved in mRNA degradation) [36, 85]. The fetuin-B deficiency must have come into account somewhere between the sperm’s capacitation, penetra- tion through the cumulus mass, the zona pellucida and the oocyte’s plasma membrane, which is essential for fertilization. In this respect, oviductal glycoproteins have been shown to bind to the zona pellucida of oocytes [86] and contribute to heightened fertilization [87]. Furthermore, they have been reported to enhance sperm capacitation, bind- ing to the zona pellucida and to facilitate sperm penetration [88], possibly 4.2. PHENOTYPING 105 by influencing the time course of acrosomal enzyme secretion or degradation [89]. Hunter et al. stressed the impact of glycoproteins on the viscosity of the luminal fluid that stabilizes the micro-environments surrounding sperm, oocyte and embryo [90]. However, the deletion of oviduct-specific glycopro- tein (OGP) did not affect female fertility [91]. Mouse models revealed that the deletion of the zona pellucida proteins ZP3 and ZP2 was associated with female infertility due to impaired follicu- logenesis, cumulus-oocyte-complex formation and the ovulation of reduced numbers of zona-free oocytes that were not fertilized [92]. However, Wassar- man et al. were able to show that mice heterozygous for the ZP3 deficiency reproduced normally despite a reduced zona pellucida thickness. Hence the thickness of the zona pellucida is not of major importance per se [93].

In vitro, fetuin-A inhibited the conversion of ZP2 to ZP2f , a process that causes zona pellucida hardening during spontaneous murine oocyte matura- tion [94]. Furthermore, fetuin-A inhibited the conversion of ZP3. ZP3 binds sperm and triggers the acrosome reaction that is essential for fertilization. ZP3 conversion causes the loss of this ability [95]. The conversion of ZP2 and ZP3 results in a block to polyspermy. Another study of Kalab et al. revealed that human follicular fluid inhibited ZP2 conversion in murine oocytes in vitro. Purified human fetuin-A was able to substitute for the follicular fluid. However, the inhibitory potential of the follicular fluid was not altered after immuno-depletion of fetuin-A. Thus other inhibitory molecules are likely to be present in the follicular fluid [96]. This result is supported by the phe- notype of fetuin-A-deficient mice, which do not reveal reproductive defects. Considering the clear phenotype of fetuin-B-deficient female mice, fetuin-B not fetuin-A might be a key player in preventing zona pellucida hardening. Fetuin-B was never studied or monitored in any of these studies and it is well possible that it was present in the fetuin-A preparations as a contaminant. The significance of the perivitelline space was revealed by Motosugi et al. in a study on oocyte polarity. These authors reported sperm swimming around the perivitelline space, possibly for further capacitation, before bind- ing to the plasma membrane. The experimental enlargement of the periv- itelline space in the non-polar body half increased the regional probability of fertilization. Based on these experiments, the authors proposed a model in which the space asymmetry exerted by the first polar body and the zona pellucida directs sperm entry preferentially to the polar body half, with no 106 CHAPTER 4. DISCUSSION need for oocyte polarity [97]. Possibly, the simple contact between sperm head and oocyte surface is not sufficient and some free space is required to trigger the fusion of sperm and oocyte. The molecular basis of sperm-oocyte fusion is still poorly understood. However, there are several animal models that reveal a fertilization blockade due to impaired sperm and oocyte fusion e.g. glycosylphosphatidylinositol- anchored proteins (GPI-APs) on the oocyte’s surface [98]. The oocyte spe- cific deletion of the Pig-a gene, encoding an enzyme involved in GPI anchor biosynthesis, demonstrated that at least one GPI-anchored protein is essen- tial for the fusion of sperm and oocyte [99]. Furthermore, the deficiency of CD9, a transmembrane protein involved in cell migration and adhesion [100] that is expressed on oocytes, was also shown to lead to impaired sperm oocyte fusion [52, 50, 51], whereas the deletion of CD81 that associates with CD9 on the cell surface showed reduced sperm oocyte fusion. The exact mechanism leading to infertility in fetuin-B-deficient mice re- mains to be determined. Furthermore, it is still unknown, whether fetuin-B contributes to fertility as a structural, or as a catalytic protein. Members of the cystatin superfamily are known to be cysteine-protease inhibitors. More- over, fetuin-A was shown to inhibit zona pellucida hardening by preventing the proteolytic cleavage of the zona pellucida proteins during in vitro matu- ration. Therefore, it is likely that the physiological role of fetuin-B is of an inhibitory nature. At this point it may be mentioned it has also been sug- gested that fetuin-B may be involved in the metastatic potential of tumors as a tumor suppressor [18]. To draw a parallel to the potential interaction of fetuin-B with the extracellular matrix of the oocyte, this might be due to the interaction of fetuin-B with the extracellular matrix of the tumor cells. A third possibility is that fetuin-B as a serum protein could be involved in transporting factors to the oocyte that are essential for fertility. Further studies are required to elucidate the molecular role of fetuin-B in oocyte fertilization.

4.2.3 Anatomy and Morphology

Fetuin-B-deficient mice were first studied by non-invasive techniques such as morphological analysis according to Fuchs et al. [101] and X-ray analysis. Morphological analysis acquires first-line information about skeletal mal- 4.2. PHENOTYPING 107 formations such as developmental patterning defects, metabolic defects, growth defects, modeling and remodeling defects, and aging and immune system de- fects [101]. Visible parameters like an abnormal gait, positioning of the limbs, or abnormal struggling, for example, would have been indicators for the conditions mentioned above. Radiography is a powerful technique for visualizing skeletal diseases. In order not to miss any skeletal phenotypes radiographs of 12 month old mice were taken using X-rays. Neither morphological nor X-ray analysis revealed any major disorders. Necropsy of the mice and subsequent histological examination of their tissue was an invasive method of searching for alterations associated with the deletion of fetuin-B. These methods revealed that the gross anatomy of the body, the organs, and the cellular morphology were virtually identical in wildtype and fetuin-B-deficient mice. In summary, the fetuin-B deficiency did not affect the anatomy or the macro and micro morphology. In particular, fetuin-B deficiency was not associated with cellular infiltrations suggesting infections or tumorigenesis. This latter point was questioned when fetuin-B was suggested to be a tumor suppressor in the literature [18]. It was thought to be involved in malign tumor progression. This is consistent with the finding that fetuin-B-deficient mice did not show spontaneous tumor formation. However, fetuin-B-deficient mice are an ideal tool for clarifying the role of fetuin-B as a tumor suppressor in an induced tumor system as has been the case in fetuin-A deficient mice [30, 31, 32].

4.2.4 Physiology and Learning

Physiological parameters were analyzed to further characterize the fetuin- B-deficient mice. To investigate manifestations of metabolic malfunctions, the weight gain of maturing fetuin-B-deficient mice and their wildtype litter- mates was recorded. However, no significant differences in the weight gain of sex- and age-matched fetuin-B-deficient and wildtype mice were discovered. Furthermore, the weight of the animals was fully consistent with the data provided by the mouse phenome database (www.phenome.jax.org). In addition, glucose tolerance tests were performed, because initial serum analysis had revealed irregular glucose levels in male fetuin-B-deficient mice 108 CHAPTER 4. DISCUSSION of the original mixed genetic background, indicating a glucose intolerance. Furthermore, fetuin-A is thought to be involved in glucose metabolism [68, 28, 29]. Glucose intolerance can be due to a deficiency in insulin production and secretion (type 1), or insulin resistance (type 2) when supra-physiological concentrations of insulin are required to achieve physiological effects. Fetuin-B-deficient mice of the fifth C57BL/6 backcrossing generation did not reveal an altered glucose tolerance when compared to their wildtype littermates. Possibly, the recorded irregular glucose levels were due to the genetic variation of the analyzed littermates: C57BL/6J mice are known to be more glucose intolerant than other mouse strains, e.g. 129/Sv mice (data provided by mouse phenome database). Since the fetuin-B-deficient mice were derived from a 129/Sv x C57BL/6 hybrid, it seems plausible that these genetic traits segregated among the littermates. Heart rate and blood pressure give insight into the condition of the cardio- vascular system. The measurement of blood pressure and heart rate at the age of 12 weeks showed no significant differences between fetuin-B-deficient mice and their wildtype littermates. This first screening implies that there is probably no cardiovascular disease to be expected in adult fetuin-B-deficient mice. As described in section 2.3.8 the blood pressure measurement method required a training period to adapt the animals to the procedure. The begin- ning of the training phase was characterized by unmeasurable blood pressure and heart rate values, since the animals were too excited about being re- strained. As a side effect of the measurements, a learning effect became obvious, expressed by the reduction of blood pressure and pulse rate. Both genotypes adapted equally well to the measurement procedure. This also indicates that the learning ability of the fetuin-B-deficient mice was not al- tered. Taken together, the physiological parameters analyzed in this study, such as weight, glucose tolerance, blood pressure and heart rate did not depend on the availability of fetuin-B.

4.2.5 Clinical Chemistry Screening

The clinical blood screen is a very powerful tool for detecting changes in various organ systems, metabolic pathways and the blood coagulation cas- 4.3. RECOMBINANT MURINE FETUIN-B 109 cade. The diagnostic impact of clinical chemical parameters for pathological conditions is well described [102] and parameter-value changes can easily be linked to clinical conditions. The clinical chemistry screening in fetuin-B-deficient mice did not reveal any manifestations of pathophysiological conditions. The parameter values determined were within the average of the control group. Again, minor dif- ferences did not reach statistical significance and might have been due to the small animal numbers that were analyzed or the variable genetic back- ground of theses animals. In support of the other testing carried out, no signs of changes in major organ systems, metabolic pathways or the coagulation system were detectable in the fetuin-B deficient mice.

4.3 Recombinant Murine Fetuin-B

4.3.1 Eukaryotic Expression of Recombinant Fetuin-B

Up to now, murine fetuin-B is not commercially available. In the course of this thesis, murine fetuin-B was expressed in eukaryotic cells, COS-7. Previ- ously, recombinant fetuin-B had been expressed in a prokaryotic system [15]. However, the eukaryotic-expression system has several advantages including the eukaryotic specific glycosylation of fetuin-B, which is absent in prokary- otes. With regard to the use of the recombinant glycoprotein fetuin-B in functional in vitro assays, the choice of the eukaryotic expression system was of major importance. Another advantage of the eukaryotic expression system was that it en- abled the secretion of the expressed fetuin-B protein. This was promoted by its endogenous signal peptide. Hence, the eukaryotic cells secreted the protein into the cell culture media, from which, subsequently, fetuin-B was purified. In comparison, the prokaryotic expression of fetuin-B had led to inclusion bodies which had to be lysed under denaturing conditions. After that, the proteins included had to be renatured. In summary, the expression of fetuin-B in eukaryotic cells yielded a much more native fetuin-B than from prokaryotic cells. 110 CHAPTER 4. DISCUSSION

4.3.2 Fetuin-B Transfecting Adenovirus

There are various ways of transfecting cultured eukaryotic cells. Depending on cell type and the intended transfection efficiency, the method of choice may include lipofection, electroporation and viral transfection [62]. The ad- vantage of viral transfection is that it leads to a homogeneously transfected cell population, due to receptor-mediated internalization of the virions. Ad- ditionally, a large number of virions deliver the recombinant DNA into one cell, thereby ensuring its rapid and plentiful expression. Furthermore the viruses may eventually be utilized in whole animal experiments to study the effect of fetuin-B over-expression in vivo. And lastly, it should be possible to rescue the phenotype of fetuin-B deficiency in vivo by adenoviral transfection of fetuin-B.

4.3.3 Inhibition of Ectopic Calcification

Fetuin-A has been unequivocally identified as a potent inhibitor of ectopic mineralization using gene-knockout technology in mice [25, 24]. This pheno- type had confirmed the results of the previously developed in vitro assay re- vealing that fetuin-A is a potent inhibitor of calcium phosphate precipitation in vitro [63, 11]. In the assay, fetuin-A inhibits the precipitation of calcium phosphate. Accordingly, fetuin-A-deficient serum has a reduced capacity to inhibit precipitation compared to serum containing fetuin-A. Fetuin-A- deficient serum has to be employed in higher concentrations to reach the inhibitory potential of wildtype serum. This indicates that there are addi- tional factors taking over the function of fetuin-A but evidently these are less effective. The question was whether fetuin-B was one of these factors. In order to analyze whether fetuin-A and fetuin-B had similar functions, fetuin-B-deficient serum was assayed for its potential to inhibit calcium and phosphate precipitation in vitro. Conversely, fetuin-B-deficient serum was able to inhibit calcium and phosphate precipitation in the in vitro assay. It did not differ from the inhibition potential of wildtype serum. Obviously, the effect of fetuin-A in the fetuin-B-deficient serum over-ruled the lack of fetuin-B. Therefore the assay was repeated with the purified recombinant, glycosylated fetuin-B. Still, fetuin-B did not show any inhibition potential at all. This is consistent with the finding that prokaryotically expressed fetuin-B was not able to inhibit calcium and phosphate precipitation in vitro either. 4.3. RECOMBINANT MURINE FETUIN-B 111

Moreover, the radiographs of 12-month-old fetuin-B-deficient mice did not reveal any signs of a calcification phenotype in vivo. Furthermore, the clinical chemistry screening did not give any evidence for interference in organical functions due to calcification. Still, these analysis have to be repeated for a DBA/2 strain deficient in fetuin-B since fetuin-A-deficient mice reveal ectopic calcification depending on the genetic background, again implying a multi- factorial predetermination. However, the results given above suggest that fetuin-B is not one of these factors.

4.3.4 TGF-β Biology

TGF-β signaling was studied because there is a short amino acid sequence within the primary protein structure of fetuin-A that is homologous to the TGF-β receptor type II. This homology was also found within the primary structure of fetuin-B as detailed in the introduction. Unpublished data of Westenfeld et al. showed that human fetuin-A interacts with TGF-β, thereby reducing TGF-β signaling in mink lung epithelia cells that were stably trans- fected with a PAI-1/luciferase reporter system. However, the interaction of human fetuin-A with TGF-β could not be reproduced employing bovine fetuin-A in this study. Bovine fetuin-A was unable to activate the PAI-1 promoter, and additionally, did not alter TGF-β signaling. On the contrary, fetuin-B was able to activate the PAI-1 promoter and further enhanced the signaling in combination with TGF-β. Fetuin-B’s activation of the PAI-1 promoter may have several reasons: firstly, fetuin-B could directly activate signaling pathways that lead to PAI- 1 expression. Secondly, fetuin-B may have stimulated the endogenous ex- pression of TGF-β that then activated the PAI-1 promoter in an autocrine fashion. This kind of stimulation would involve de novo protein synthesis and would be time dependent. Therefore it could be constrained by shorter assay times or the inhibition of protein synthesis. Thirdly, either TGF-β or other substances that can already cause the activation of PAI-1 in low amounts may have contaminated the purified fetuin-B preparations. This is a known problem in virus-derived proteins. To exclude this possibility, fetuin-B should be expressed in a virus-free system. Thus, a direct answer as to whether fetuin-B interacts with TGF-β ex 112 CHAPTER 4. DISCUSSION vivo could not be given by this experiment and more refined experiments are required to clarify this point.

4.4 Outlook

The infertility of the fetuin-B-deficient mice opens a novel research field for fetuins. This work describes a novel target for the investigation of idiopathic sterility and for birth control. The future aim will be to elucidate the molecular mechanism in which fetuin-B is involved in successful reproduction. In this respect, in vitro fer- tilization (IVF) studies will provide information regarding the sperm’s ca- pacitation potential that might be reduced in vivo in mice lacking fetuin-B. Furthermore, by removing the zona pellucida, IVF studies will be able to elucidate the contribution of the ZP to the fertilization defect. If IVF studies fail, further intracytoplasmic sperm injection (ICSI) will provide information about the developmental potential of fetuin-B-deficient oocytes. Later stages of pregnancy may also be affected by fetuin-B deficiency. Thus, embryo-implantation studies are planned to demonstrate whether ma- ternal fetuin-B is essential for later stages of pregnancy, such as implantation and the maintenance of pregnancy. Summary

This study aimed at elucidating the physiological function of fetuin-B. For the first time, the serum protein fetuin-B was deleted in an animal model. This was achieved by gene targeting in embryonic stem cells. The analysis of the fetuin-B-deficient mice revealed that fetuin-B was essential for fertilization. Female fetuin-B-deficient mice failed to produce any offspring when mated with any genotype. Heterozygous fetuin-B-deficient female mice and males of any genotype were reproductive, however. The main finding of this study was that oocytes derived from naturally mated fetuin-B-deficient female mice did not reveal any fertilization characteristics and did not develop to two-cell pre- implantation embryos in vitro, indicating a fertilization defect. It remains to be clarified whether fetuin-B is required as a structural, a catalytic, or a transport protein. Regardless of the exact mechanism, a novel target for fertilization control was identified: fetuin-B.

Further characterization of the fetuin-B-deficient mice did not reveal any abnormalities with regard to their anatomy, morphology, physiological pa- rameters or metabolic functions. It remains to be studied if the fetuin-B- deficient mice develop disease when challenged or stressed. For example, the hypothesis that fetuin-B may function as a tumor suppressor preventing tu- mor progression could ideally be studied in an induced-tumor model using fetuin-B-deficient mice.

Functional in vitro assays performed with purified recombinant fetuin- B showed that it did not compensate for fetuin-A deficiency, as shown by their distinct calcium phosphate precipitation inhibiting potential. As for the fetuin-A- and fetuin-B-deficient animal models, both exhibit distinct pheno- types and differ completely in their most prominent phenotypes: fetuin-A deficiency leads to ectopic calcification, while fetuin-B deficiency leads to fe-

113 114 SUMMARY male infertility. The data provided by this study thus clearly suggests that the fetuin-family members have distinct functions in vivo. Appendix

Pedigrees

1/2D2(FetA-/-) x 3+/- (17.08.05 - 19.12.05)

N1 1.1 SPF 1.2 SPF 1.3 SPF 1.4 SPF 1.5 SPF 1.6 SPF 12.09.05 19.09.05 15.10.05 24.10.05 12.11.05 27.12.05 - 5f 2m 4f 4m 2f 3m 1f 6m 3f - 3f 9/10+/-(FetA+/-) x 15D2(FetA-/-) (28.10.05 - 11.01.06)

2.1 SPF 2.2 SPF N2 23.11.05 19.12.05 12m 5f 8m 9f 57/58+/-(FetA+/-) x 15D2(FetA-/-) (07.02.06 - 27.03.06)

3.1 SPF 3.2 SPF 3.3 SPF N3 28.02.06 30.03.06 04.04.06 4m 10f 6m 3f 5m 6f 74+/- x 106D2 (05.05.06 - 19.07.06)

4.1 SPF 4.2 SPF N4 30.05.06 28.06.06 4m 4f 5m 5f 109/110+/- x 106D2 (19.07.06 - 23.11.06) 5.1 SPF 5.2 SPF 5.3 SPF 5.4 SPF N5 02.09.06 25.09.06 22.10.06 30.10.06 - 5f 2m 4f 4m 2f 3m 1f 125/133+/- x 106D2 (30.11.06 -) 6.1 SPF 6.2 SPF 6.3 SPF N6 11.01.07 21.02.07 01.03.07 3m 4f 3m - 3m 4f

Figure 4.1: DBA/2 backcrossing pedigree. Beginning from the second C57BL/6 backcrossing generation as shown in Figure 4.2, this pedigree shows every DBA/2 mating event. Litters are indicated by a box, showing date of birth and numbers of male (m) and female (f) pups. Number code: the first digit indicates the mating cage; the second digit counts for the litter number of the mating cage. N = back crossing generation.

115

116 APPENDIX

N6

N5

N4

N

N3

N1

N1

N8

N9

N7

2

(07.0

128B6 x 190+/- x 128B6

1m

26.03.0

9.2 SPF 9.2

2.06 - 26.05.06) 26.05.06) - 2.06

3f 3f

6

2m

18.04.0

9.3 SPF 9.3

F2

F1

0f 0f

6

(05.09.06 - 30.11.06) - (05.09.06

225/226B6 x 261+/ x 225/226B6

23.

2m

Emb

2m

25.07.06

18.1. SPF 18.1.

04.

3

Bonn/

m

0m

1

12.05.0

9.4 SPF 9.4

3.

0.2004

0

C

1

1.

3f

3f

h

3f

0

1

5

imer

2f 2f

6

ryot

1m

18.08

18.2. SPF 18.2.

X

2m

14

a

3

.01.

B

4

3

3m

.

26.03.06

8.1 SPF 8.1

2

.06

: :

: f :

1f

2

f3.1

0

2

f

r

-

5

6

.1

an

22.10.05

135/136+

.1 x m2.1 .1

.1/

0f 0f

(09.0

226B6 x 226B6

(05.09.06 - 30.11.06) - (05.09.06

225/226B6 x 324+/- x 225/226B6

f1

6m

11.09.06

19.1. 19.1.

2

04

m

.1.3 x m2.1 .1.3

sf

3

.02

6

4m

2

5

19.04.06

8.2 SPF 8.2

1

.3

3.06 - 26.5.06) 26.5.06) - 3.06

m

8

.1

.1

S

6

.11.05

.3

.

6f

e

0

PF

- 22.11.05) 22.11.05) -

f

S

S

/- x 149B x /-

209+/- 209+/-

5

(07.02.06 - 24.05.06) 24.05.06) - (07.02.06

127B6 x 189+/- x 127B6

PF

PF

r

4

5f 5f

:

f

8m

.1

25.10.06

19.2. SPF 19.2.

1

-

119B6 x 1 x 119B6

9

5m

14.05.0

8.3 SPF 8.3

0m

3

.02.

11.

12.3 S 12.3

6

4 4

20

5

1 1

.4

07.08. 07.08.

m

m m

.2 .2

.2 .2

1f

6

.12

1f

05.0

SPF SPF

0

SPF

5

3f 3f

5

.05

4f

P

2f

2f2f

6

05 05

6

F

5m

2

ma

Bo

6

2

21.

.

18+/- 18+/-

m

1

nn

0

3

2

3 3

1.1.

0

5f

.04

.5

31.

/2

24. 24.

m

m m

2

1

4m

8m

1

4 SPF SPF 44

3m

5m

14.07.06

11.

4

04

7

12.01.07

20.2 S 20.2

04.04.

16.1

IVC+

9

.05

m

4

.

.

f

0

4

1

10. 10.

.10

.

l

(01.0

3

1

0

S

SP

e

.05

SPF

1

(09.03.06 - 26.5.06) 26.5.06) - (09.03.06

225B6 225B6

SPF

1f1f

05 05

(19.03.07)

373/374+/- x x 373/374+/-

PF

(30.11

225B6 x 324+/- x 225B6

.05

11f

.06

(22.06.06 - 14. - (22.06.06

255+/- x 149B6 x 255+/-

(14.12.05 - 01.06. - (14.12.05

151/152+/- x 149B6 x 151/152+/-

1f

3

06

2f

3f

PF

F

6:

5: f 5:

f

5m

10

(-/

6.05 - 16.08.05) - 6.05

f3

3

4

.01.

.

.1

.

1.

3 3

3m

8

1

23.

20.09.06

16.2 SP 16.2

3m

14.02.06

7.2

4.

26.03.07

20.3 SPF 20.3

7

x208+/-

.06 -15.0 .06

.2/f3

1 1 m m

2 2

-

m

4

3f

m

2.2.

0

7 7

5

)

1

5

x

. .

11

0 0

SP

S

1

(17.08

m1

127/128B6 x 123+/- x 127/128B6

.1.3 x m1.1.2 .1.3

6.05 6.05

SP SP

PF

.05

0f

5f

4f

5f

F

x

F

.

f f

8m

F F

1

377B6

04

.4

12.06)

4

.02.

3.07)

1m

23.11.06

16.5 SPF 16.5

.2

3 3

.0

2m

B6

30.03.06

7.4 S 7.4

4.

3

05.

1 1 m m

07. 07.

4f

m

2 2

6

5 - 22 - 5

0

06) 06)

.2 .2

5

S

1

0 0

SPF SPF

2

PF

PF

8 8

(1

2f

2f

3f 3f

.05

.05 .05

7.08.0515.11.05) -

f f

10m

13

.

11

5.

.01.

3m

15.12.06

16.6 16.6

121+/- x 122+/- 122+/- x 121+/-

2m

si

1

wurd

1

21.04.06

7.5 S 7.5

.

1

05

m KO a KO m

4.10

b

2

0

l

f

i

n

5

)

SPF

e

g

P

4f

.2005, .2005,

t

s

1

1f 1f

F

o

),

8.10

Bo

t

m 01. m

4

i

B

08.

m

m m

l

nn

ut

5

.04

ei

02

K

4m

11.05.06

7.6 S 7.6

.2

zuck

4f

/3

n ä

0

3f

.

e

f

7.200

0

i

g g g

r

5

e

davon i davon

P

N3/F2

rwe

4f 4f

4

4m

F

e

m

07

07

f

3

6.

5

unde

6

.1

sta

rt

17

1

. m

.09

e

0

SPF

S

5

.02.

st

9

1m

12.05.06

7.7

b

r

PF

.3

.05

.

n

3

k

ei

3f

0

seh

, ,

ve

f

3

5

0

st

f

al

S

5

a

r

PF

le

r

k

f

r

0f 0f

k

r

3

n

3

et

20

atzt atzt

3.

04.

6

m

una

m

v

si

x

t

.2

2

2

e

1

.10.

(4

m

0

rwe

SPF

0.

SP

e

m

1.06

6

1

u

288/297+/- x 291+/- x 288/297+/-

286/287+/- x x 286/287+/-

275/285+/- x 283+/- x 275/285+/-

271/273+

248/250+

246/247+

207/227+

04.0

01.05

3

he

D

.1

g

i

05

ff

2

t Ha

F

f

st

s

ä

f

3

,

l

t

f

li

att

BA

g

D

dr

7.06 Kol 7.06

utlä

.

30

e

6.4 S 6.4

B

2m

28.01.

i

si

l

A

eben no eben

2m

o

g

09.

/- x 281+/- (S1-VK4) 281+/- x /-

/- x 280+/- x /-

/- x 243+/- (S 243+/- x /-

/- x 237+/- (S1-VK1) 237+/- x /-

n

/2 /2 /2

b

P

en,

6

2f

e

0

F

06

.2

i

2

oniewechsel von: oniewechsel

S

d

284+/- (S1-VK6) 284+/-

e

3f

.05

en en

ta

i

ch a ch

ner

6.6 S 6.6

3m

m

18.03.06

m m

m

(S1-VK7)

(S1-VK5)

(S1-VK3

P

b

1f

F

1-VK2)

aum

st

sie

)

amm

h

sieheS1-S

e Homozy e

ba

um

gotenverpaarungs-

t

a

mmb aum

Figure 4.2: C57BL/6 back crossing pedigree. Beginning from the generated chimera, the pedigree shows every mating event. Litters are indicated by a box, showing date of birth and numbers of male (m) and female (f) pups. Number code: the first digit indicates the mating cage; the second digit counts for the litter number of the mating cage. N = back crossing generation. FETUB GENOMIC SEQUENCE 117

Fetub Genomic Sequence

>Nt_039624contig Mus musculus chromosome 16 genomic contig, strain c57BL/6J. gtgaccagattgacttctgatgatttcacagctattcacacggaattagaaaggatacct gctttgtgctaacacggctataaaacaggatgacgggacctttgatgcagccgccctcct ctccattccacgtgagcgtcctggacttacccatgcccagcctgctgcgggcagccagag cctttctgggtcactgtgtgtacagtactgactatggtgagattcagcaataaagccacc tactgagtgacagccttgggcatcagcagctctgggtggagaaggccctgtgcagggaag ccacagcctatgggtgggacccaaagtgcgggcagagcattgggaaaagcccaggcagaa acaaagtactgggcggctccttggaaagacacggaacctaattagagcttattctctcta ggaccttcctctaattcatagttagggtgtatgtcagccctgaaactcagctaagcaggg tattattcctggagttctgaatgatggcgcactctgtttgcaagatataaaagggcagtc agagggagaagctgccatcatcctttgcagctccatcgggaacacctggagaggatCCCT CCAATTTGTTAATTAACAAACAAGCACCTTGTTTATACACCAGTTGCTCCTGTGAGTTGG TCTCCTGGCCGAAGCGACTCTGGGTCCTCAGGAAAGCCTGTCTGTCAGAGCTGCAGGACT GGGCCCCACCTTCCGTGTTCCTCCCTGTCTTAGATCAACGGGACTgtgagtatgtgacat gcgtggccatcttccctgcccactccttactttttagactcttttggggggggggggagt acagaggccaaatccatagccaagcacagttcactggtcctcacacccagcccacgtgtg tgtggaaaatgcttcctgcattcccattggtgagtactgaactggaattaaaccccaggc ctgcgtgaacccagcgtctgtattagtggcctcttcacaacaaggatttttacgggaaat ctcactcatgatgcaataggatcacgttcccccgggggggcatggtggatagatctgtgt gattcaagttccggaaatctcactcatgatgcaataggatcacgttcccccggggggcat ggtggatagatctgtgtgattcaagttccgtagagccaggttctaggagctgcccctgct agagatggaatggcaccctgtgcagagaggggggcagagtgccctcccatcagggctaaa cgggcccttagtaatcgcatggttgaaagccctacacagttgaggaaattgggcttaagt gttacatcgctagcctagctcacgcagaccagggctgaagcagggtcctggtcaaagttc agtttaccatgtgatttagcatccttacagcatctttttaccccgtttgcatctgtcttt cattaaaattgcaccaatggcttctacctaagtgtactgggtacatttcctcaggcattg gacactccagcccagacccgacagaggtatgttcactgctggctgaaactaaatctctgc tgaacctcagagtttggatatctgattttttttaactgataaaggaaaacttaagcacat ttaaattgaaaaaaattaaagcattcattaatttatgagtcccagtattgtgggagggta aacttgtacaatttagctgtagaaagaagggaagataaatgaattttattggttagtgta gaaagctcctgtcttgaggttattatttatttgtttgttgtttgtttatttggtagagat ttaaattccagagcctccacaatctaatggtctcccaatttgtgttgtttttcttaaagt agccatttctggcttcgatacccatttttctcgaattattcccatttgaaaacacaactg ggagcgtgtgggggaaaaaaaatttggtaactcagggttggaatcattctaaacaacacg tggcatgagccaacccagaaaaacaggtctttaaagatatttctaaattggaaaaaagaa agaaagaaagaaagaaagaaagaaagaaagaaagaaagaaagaaaacacaaccaagagct cccttctgtagaactgtaatcttcagatttgggtttcataatcaatctttactaaccact tgtttacggaagactaaaaagtgggcgtggcttcttattaatgtggtccatcctgtgctg tgctgctgtcaccggatccttctcacccttctggaggcctggagggcctctgggaacagc tttcttgctgcgtcatgcgtgggcgggaggcgggaggcaggaggaagaactctgcctaac aaatgtactcccctgacatgggcatgccccacccctgagaacagagcccttgggacttaa tctctgtaaaggcgccacctctccacaggtttcataagcgttcgttttctgagctttggg aacgtgttcaaagcaCACCAGACTTGCAAAGCTCTTGATTCCGGACAGCTTACCTTGATT CCTGACATTGTGCAGTGGCTCAGCCAGAACCTGTGAGTCACAGGTAGCTGCTGTGTTACA CAACATCTAACAACGTCTAGCCTTCTGCGATTTCTGGTGCAGGGTCATGATTAGTGCATT TGACAGATTTGGCAGAGTGGCTTACAGTGGgtaagtggcttctccacatcggctggttgg tagctgaaagagtagctcacacagtgttctaagcatctgtgtggtgacaaaaatattttt acatttatttgtttatttttgtgcagaggtggggtgggtgggccagagtgcagatgacac tgcactctggtcagaggacatcgttcagcagtcggttcttccctttcactgtgagggtat caggagttacattcaggccaccaggcttggcagcaagcacctttaggggctgagccatct cactcgctttgatgaaaatcttctaaaagggaaggctcctgtgctgtttcttgtattgat tcattcaggatttttaccctttctgagccctagtattctcccctggttggacttggctaa gttttctttagagttttcagtgagtgtctatactaccactaagttagtgagtggatgctg ttacctagttgtctatgagtagctatcaaatggggtggggggtggggtgggggggaagga atctgtgttctgctcaactataatacaattacatttagcatcataaatttaagtttttta gtaagtttattataaaattaacttttcaatgttagctaaaaacataagtcagcatattta agtttaatatgtaatcaatgttttaagtaaatttaggaacttgagcaatgttaacagcct tcttttgcttggttcactcgtgtcagccatagcttactgcatattggattttcatctaga ccagaggagcatttctttccatccaaaatatttgggccctcttatcaggttcccatgaaa agtctcacagactggtgtccttatttataaacacagacatttccaaaatatatttgtgaa ttaaaaataagctctactcaggaaaactcttatattctctagtagctgtagggacctgtg 118 APPENDIX

tttgaaatagtattgaccccaaagctttggagtcttcaagatactctgtgtattgagggc ctaccagatgggtacctgtggatagagcagaacctctgtctattgaacacagaaagcctg ggcaggggtggagatggaggtatggaatgaggtattgcttaccttagggaatgctttaag ggacagcctgctatgtaggctcccttcctgacttagacctttgctttcattgttgatatc tctctctctctctctctctctctctctctctctctctctctctctctctcccccccccca cccccgtgtgtgtgtgtttgtagtagaaacatttttaaaaaaataaatggcctgtggctg tagctcagaagtacaatgtttgcctggtgtaagtgaggccctggtttgatgcttggcatc atgcacaatgtgtgtgtgccatttatggaggtaggtacccgcaatcccaggaatttgagc agctgaagcaggattgcaagttccatgccagactgggaaacttattgaggtcttgtctca aaataaaagttgctgtgatgtagcgctatgaaagcacgtccccagcacatatgaggccca gccctagagggaaaggagctttgaaagatacctgatttagtcaccaggtatggtgatatg cacctgtagtcccagtagatgcaagaggattgcgagttcaaggccagctaccgaacacat ttaaggccctgtcaaaacaaaaaataaattgcttttcatatatccccaataagcagaacc ttcctgggatctgaactgaagctcagaaagagaggcaagaaccatgatccgtcttgatgg ccaccattatctttctcttgacagtgtcaggtcttggtctcttgatgaatttacctgtag gtgcacaaaaggaagactagacctagagtgtgttgggagaacttttggggtctccttagc tttccccaccttaaatacaattctgagatgaagcattttcctgtcctatacaggaactag aaagggacacttctgagtagtgcttgtattcagttccagttggattaggactctccatct ggacaagacagaagccaggtctcccatgtcttgatcaaacatttttttataattcctggg aaagggttaggaaacttggatttatttacacgaaaggcatgctccggatccaaggttaga acactgacaaatccaatccaaaatattgactcaaaaactgctgtgtttggatagaggtgg cctcagggcatggtgtttcaagtcctagtaggctattctgttgcagttataaagctgctc aagtgaaccttgaggccccacggcagggctcgtcctggcagctctatactgacacatcct ggatctttttttttttttttttttttttttttccgagtttgttttttttttttttttttt tttttccattttttattaggtatttaactcatttacatttccaatgctataccaaaagtc ccccatatccacccacccccactcccctgcccacccactccccctttttggccctggtat tcccctgtactggggcatataaagtttgcaagtccaatgggcctctctttccagtgatgg ccgactaggccatcttttgatatatatgcagctagagtcaagagctccggggtactggtt agctcataatgttgttccacctatagggttgcagatccctttagctccttggctactttc tctagctcctccattgggagccctatgatccatccattagctgactgtgagcatccactt ctgtgtttgctgggccccggcatagtctcacaagagacagctacatctgcgtcctttcta taaaatcttgctagtgtatgcaatggtgtcagcgtttggatgctgattatggggtggatc cctggctatggcagtctctacatggtccatcctttcatctcagctccaaactccgtctct gtaactccttccatgggtgttttgttcccaaatctaaggaggggcatagtgtccacactt cagtcttcattctccttgagtttcatgtgtttagcaaattatatcttatatcttgggtat cctaggtttggggctaatatccacttatcagtgaatacatattgtgtgagtttctttgtg aatgtgttacctcactcaggatgatgccctccaggtccatccatttggctaggaatttca taaattcattctttttaatagctgagtagtactccattgtgtagatgtaccacattttct gtatccattcctctgttgaggggcatctaggttctttccagcttctggctattataaata aggctgctatgaacatagtggagcatgtgtccttcttacctgttggggcatcttctggat atatgcccaggagaggtattgctggatcctccggtagtactatgtccagttttctgagga accgccagactgatttccagagtggttgtacaagcctgcactcccaccaacaatggagga gtgttcctctttctccacatcctcgccagcatctgctgtcacctgaatttttgatcttag ccattctgactggtgtgaggtggaatctcagggttgttttgatttgcatttccctgatga ttaaggatgttgaacattttttcaggtgcttctctgccattcggtattcctcaggtgaga attctttgttcagttctgagccccattttttaatggggttatttgattttctgaagtcca ccttcttgagttctttatatatgttggatattagtcccctatctgatttaggataggtaa agatcctttcccaatctgttggtggtctttttgtcttattgacggtgtcttttgccttgc agaaactttggagtttcattaggtcccatttgtcgattctcgatcttacagcacaagcca ttgctgttctgttcaggaatttttcccctgtgcctatatcttcaaggcttttccccactt tctcctctataagtttcagtgtctctggttttatgtgaagttccttgatccacttagatt tgaccttagtacaaggagataagtatggatcgattcgcattcttctacatgataacaacc agttgtgccagcaccaattgttgaaaatgctgtctttcttccactggatggttttagctc ccttgtcgaagatcaagtgaccataggtgtgtgggttcatttctggatcttcaattctat tccattggtctacttgtctgtctctataccagtaccatgcagtttttatcacaattgctc tgtagtaaagctttaggtctggcatggtgattccgccagaagttcttttatccttgagaa gactttttgctatcctaggttttttgttattccagacaaatttgcaaattgctccttcca attcgttgaagaattgagttggaattttgatggggattgcattgaatctgtagattgctt ttggcaagatagccatttttacaatgttgatcctgccaatccatgagcatgggagatctt tccatcttctgagatcttctttaatttctttcttcagagatttgaagtttttatcataca gatctttcacctccttagttagagtcacgccaagatattttatattatttgtgactattg agaagggtgttgtttccctaatttctttctcagcctgtttattctttgtatagagaaagg ccattgacttgtttgagtttattttatatccagctacttcaccgaagctgtttatcaggt FETUB GENOMIC SEQUENCE 119

ttaggagttctctggtagaatttttagggtcacttatatatactatcatatcatctgcaa aaagtgatattttgacttcctcttttccaatttgtatccccttgatctccttttgttgtc gaattgctctggctaatacttcaagtactatgttgaaaaggtagggagaaagtgggcagc cttgtctagtccctgattttagtgggattgcttccagcttctctccatttactttgatgt tggctactggtttgctgtagattgcttttatcatgtttaggtatgggccttgaattcctg atctttccaaaacttttatcatgaatgggtgttggatcttgtcaaatgctttttctgcat ctaacgagatgatcatgtggtttttgtctttgagtttgtttatataatggattacattga tggattttcatatattaaaccatccctgcatccctggaataaaacctacttggtcaggat ggatgattgctttaatgtgttcttggattcggttagcgagaattttattgaggatttttg catcgatattcataagagaaattggtctgaagttctctatctttgttggatctttctgtg gtttaggtatcagagtaatagtggcttcataaaatgagttgggtagagtaccttctactt ctattttgtgaaatagtttgtgcagaattggaattagatcttctttgaaggtctgataga actctgcactaaacccatctggtcctgggctttttttggttgggagactattaataactg cttctatttctttaggtgatatgggactgtttagatggtcaacttgatcctgattcaact ttggtacctggtatctgtccagaaatttgtccatttcgtccaggttttccagttttgttg agtatagccttttgtagaaggatctgatggtgttttggatttcttcaggatctgttgtta tgtctcccttttcatttctgattttgttaattaggattttgtccctgtgccctttagtga gtctagctaagggtttatctatcttgttgattttctcaaagaaccaactcctcgtttggt taattctttgaatagttcttcttgtttccacttggttgatttcactcctgagtttgatta tttcctgccgtctactcctcttgggtgaatttgcttcctttttttctagggcttttagat gtgttgtcaagctgctagtatgtgctgtctcccgtttcttcttggaggcactcagcgcta tgagtttccctcttagaaatgctttcattgtgtcccataggtttgggtacgttgtggctt cattttcattaaactctaaaaagtctttaatttctttctttattccttccttgaccaagg tatcattgagaagagtgttattcagtttccacgtgaatgttggctttccattatttatgt tgttattgaagatcagtcttaggccatggtggtctgataggatacatgggacaatttcaa tatttttgtatctattgaggcctgttttgtgaccaattatatggtcaattttggagaagg tcccgtgaggtgctgagaagaaggtatatccttttgttttaggataaaatgttctgtaga tatctgtcaggtccatttgtttcataacttctgttagtttcactgtgtccctgtttagtt tctgtttccacgatctgtcctttgaagaaagtggtgtgttgaagtctcccactattattg tgtgaggtgcaatgtatgctttgagctttactaaagtgtctctaatgaatgtggctgccc ttgcatttggtgcgtagatattcagaattgagagttcctcttggaggattttacctttga tgagtatgaagtgtccctccttgtcttttttgataactttgggttggaagtcgattttat ccgatattaaaatggctactccagcttgtttcttcagtccatttgcttggaaaattgttt tccagcctttcactctgaggtagtgtctgtctttttccctgagatgggtttcctgtaagc agcagaatgttgggtcctgtttgtgtagccagtctgttagtctatgtctttttattgggg aattgagtccattgatattaagagatattaaggaaaagtaattgttgctcccttttattt ttgttgttagagttggcattctgttcttgtggctgtcttctttttggtttgttgaatgat tactttcttggttgttctagggcgtgatttccgtccttgtattgcttcttttctgttatt atcctttgaagggctggattcgtggaaagatattgtgtgaacttggttttgtcgtggaat actttggtttctccatctatggtaattgagagtttggccgggtatagtagcctgggctgg catttgtgttctcttagtgtctgtataacatctgtccaggctcttctggctttcatagtc tctggtgaaaagtctggtgtaattctgataggccttcctttatatgttacttgacctttc tcccttactgcttttaatattctatctttatttagtgcatttgttgttctgattattatg tgtcgggaggaatttcttttctggtccagtctatttggagttctgtatgcttcttgtatg atcatgggcatctctttttttatgtttgggaagttttcttctattattttgttgaagata ttagctggccctttaagttgaaaatcttcattctcatcaattcctattatccgtaggttt ggtcttctcattgtgtcctggattacctggatgttttgagttaggatccttttgcatttt gtattttctttgactgttgtgtcgatgttctctatggaatcttctgcacctgagattctc tcttccatttcttgtattctgttgctgatgctcgcatctatggttccagatctctttcct agggtttctatctccagcgttgcctcgctttgggttttctttattgtgtctacttcccct tttagttctagtatggttttgttcatttccatcacctgtttggctgtgttttcctgcttt tctttaagagcctgtaactctttagcagtgctctcctgtaaatctttaagtgacttatga aagtccttcttgatgtcctctatcatcatcatgagaaatgtttttaaatctgggtctaga ttttcggttgtgttggggtgcccaggactaggtggggtgggagtgctgcgttctgatgat ggtgagtggtcttgatttctgttagtaggattcttacgtttgcctttcgccatctggtaa tctctgaagctagctgttttagttgtcactgttaagagcttgttcttcaggtgactctgt tagcctctatgagcagacctggagggtagcactctccttagtttcagtgggcagagtatt ctctgcaggcaagctctcttcttgcaaggcaggtacccagatatctggtgttcgaaccag actcctggcagaagttgtgttccactcactagaggtcttagggtcccgtggggagtcccg tgtggacccttgcgggtgttgggcaagactctgctggcaaggtagcccggggctcgagtc tcgagtcgagcggaggggacttgtgccccagatcaggcccgggtagcctgcttccctatg taccgcagtctcaagttccgcgcgattggattggggcaggcactgtggtccactcaccag aggtcttagggtcccgtggggagtcccgtgtggacccttgcgggtgttgggcaagactct 120 APPENDIX

gctggcaaggtagcccggggctcgagtctcgagtcgagcggaagggacttgtgccccaga tcccacatcctggatcttgttccccacagAATGGGCCTGCTCCGACTTCTGGTGCTCTGC M G L L R L L V L C ACCCTGGCTGCATGCTGCATGGCCCGCTCTCCCCCAGCCCCACCCCTGCCACAACGCCCC T L A A C C M A R S P P A P P L P Q R P CTCTCACCTCTGCATCCTCTGGGCTGTAACGACTCTGAAGTGCTGGCAGTTGCAGGATTT L S P L H P L G C N D S E V L A V A G F GCCCTGCAGAACATCAACAGAGACCAAAAGGATGGCTATATGTTGAGCCTTAACAGAGTG A L Q N I N R D Q K D G Y M L S L N R V CATGATGTTCGGGAGCACTACCAGgcaagtgacaggttgtggggcagcatttgggcaggc H D V R E H Y Q tgcaggcactagccagggggtggcaacttggtgctacttgtcaacaggtactttctggtc tagaaacagaatactctgccgtcaccagcaggtgtgctatgacactcttgctggccatag gaatggtgacaagagtgtttctactctgtcccaggtccaggcttcaggagtatagcagag catgtatcacacacaggagcttaggttctcccaggaagatgctattggtctagttgtcct ggatctgtttcatgtgcaatgccgtatttctctaacccttatcatagGAAGACATGGGAT E D M G S CTCTGTTCTACCTCACATTGGATGTCTTAGAGACTGACTGCCACGTGCTCAGCAGGAAGG L F Y L T L D V L E T D C H V L S R K A CACAGAAGGACTGCAAACCGAGGATGTTCTATGAGTCGgtaagtgcctgttcccaggggc Q K D C K P R M F Y E S ctgctcagtgtctaccgtctgtttcatcttcattatctcccaggcaacaggccagtgtgc caattctaatactggaggaagagccaagtttagtcagggtagggaggggtcctcagactg gggccttgctgactagacttgaaaagtttcccccaaattattttctaccctaagaatctt tggcagctgagccaatgtttatagtctcgggggggggggggcaaagacagagattgatta attgattgattgattgatttctggtgtttgaggaaggtaggtagacagcagcaataatgg agtatagcttgtgttcttttgtttggttttatttgaatgaatatcaatatgaggttttca gataggaaagaggacagctttcagtgtgggtactgtcaggggcttcctgggcataaaact gaacggcctcagaactcatcctttctccttttaaaaaaatattttatttttctatacata tgggtgccttgtgtgcatgtatgtgtatgcaccacgttcgtgcccacagcgaccagaggg tggcaaaagacccaccccaccccccaccccaatctccaaccctagtactggagtttcaga tggttatatgccacctaatgggggcactgagaaccaaacttgggttgtctagaagaatcg tgagggtcactaagccatctttccagccttgagcttggcctttcaaaatggccactggca agacttgaaaccagtgcagtcagcttccacctgactctcagtgattctgggatctctttc attcactgatgcatggaaggtgtatgacatggaagacaaggtgagatcttcagtggtttg aagtaaactttttgatcaagatcaccttttgtagttatttgaccaaacattccgacctgc acctgttttttgtttgtttgttttgtttttaaagcaaaaacaaacaaacaaacaaaaaac aaagaacaaaaaacaaaacaaaacaaatcccccaaaccaaaaaccaaataatagaaaaac cagtcttatgccttcttgttatagttggtttagtgatacgggccactcacagagatatgt atgctcactcagggacacacaggttaggagtttatttcacgtgtaatttcaaacttcaag ttcctgaccaggggattttgctctgggcatgatttggaggccccacttctcgctgtgtgt tgggcaccaggcgtgggggaataaagggagtgggagaaaaccccgcatcccaccagagtt ccggttctctgggtaggcagacatgggaggactgctgcatgctttccacatggccccggg tgggctgtgggaagctatggacccagcccagggggatgggcaaggggcattccaaggtct ctgggcctcatggaggccagagataacaggttgtcaggcatcccagacaccactggacac cgcagaagagctgagaacaaaatgggtgcccctggggctgctcggtcagccctgggcccg aagggaggagagggcaggaggagagggggtactgggtggttcccatgccggtaagagtct ttggtctggtccatggctggagctcaggaagaccttcgggcggggttaggggaaagccta tcccatcgttcaagcactgcaggcctgatgaacagagatagtctatggttttagagattt attatagaaaggcaaggggaaagagagagagagaggctggccatggccacgtggagagag gggcttagtgagagagagaaggagggctagagatgagagtaagaaaggcaagagcttaga gagaggaggggccaagcagttccttctatagtgggctgggctaccttgctgttgccaggt aactgtggggaggagcaaacctggctataggttaactttgggggtggagtctagccagaa tgttaggagcttggggctttatctgtgtgactgatagtcacagaattatggagttgggcg ctctgcggtgtcaggcacctgtctctgggaatgtggctcagtgttctgcagagttttcta ctgggacgtaggagcaagcttcattcaatcaaaacaggctgcctttcacggccccacatc tgtgcttgtctctgaccttgctacctcctacctgtgagtccctagtgcccatattcactg gcatcaggatagaacgaggacatgccatgctgacgtggccatggagcagggcaaagggaa gctggttctgatccatttctgcagttagaactcaggttggcgatccacgcccgagtgtga ggagcctgggaaggtagctgtgtcccaggaaaacatggaagagtctgatgctaaagagtt ctccccaccccaaacacagaactggatttaagtttcacattacattaaaatacaattcta cttgggggaggggtggtgtttaacctcgtggttctctgcagcatgccatcccatcgagta FETUB GENOMIC SEQUENCE 121

catttgttttttgttttttcctagGTTTATGGCCAGTGCAAAGCAATGTTTCACATTAAC V Y G Q C K A M F H I N AAGCCAAGAAGAGTTCTCTACTTACCTGCTTATAACTGTACACTTCGCCCAGgtaaacat K P R R V L Y L P A Y N C T L R P V caagacggttttgtacatcttgtcatcaaaaaacagaaaagagaacttaactagggcctt tgatgagttggcaagcaatctctcctctcttccgctgtatccatcacagcactgcacaac actccatcattgcctctctactgacattccctccatacttctcctcccaggggacagcct tcctaagtcactgtcaagagctctgtcctgatccagatcctagatgctcattcttctgtg agtgacacaatagcaatagctggtcccaggatcccttctatctgtcccactgtcctgaca cccttccagctggctgttgtgttgtcagaacacgcactacactttcctgtgtctgcactt gtttgagcctctccctctgctgtatgcccttgatggcctctgtatcagaactcatacccc cttttatttaaaacaaaaaacaaaaaaacacaatgtttttgagatgccctttactcaaga aaggaatggttctttcctggtgtaaatttttctcactttggctaaaacactcatccaatg cagaacagaacatgcacaatcttgacttatagtcttatatcttctatcatatggcatgca tgtccctctgccccagtaaaagacttcctaatttatataaagttttggaggttattacca ttgctgaataacaggggaaaaataaggctggaatgaataggccattcattgaaataagtg tgtgcactcttccctctggagcctgctttttacaagagagggtcttggaaaagtgggagc ccattgcacgggaaaatgggaaatatctacctagcagcttgtgctacctcactgtgataa aggggccagcctgaaatcaatctggggacttctggaaatggtggcagtgatacccatagg tgtcaggagttggtgacgagggaggcacagtggcaggctgtgggcacagctctcccgcca tcttcctccacctccaaacatttattagggaagacaatgaattttctgacaacaccccgg ggcaaattccctggcttgatgagcacacatttgtcctctccagctcctcctgtcatggtg tcatccacttgcttgacaggcctctactggctcatcagtctgtgttaggtttgctcgtgg tgctgaggaccaaggccatgaagtagctcctgacaataatgaagtgtctatgggaacagt gatgggaaaggacctggagctagggaggggactgatgtgtctactgttttgacacaggaa cacaatgctgtcatctaccatgttcacacttgataactgtgtaatcatgttgcaaaaaaa aaatctcgtaatgttttaagcatatttacaatcccatgataggtcgcattctttgtggtc tttaacactgtgctgtccacagactactggttggccatgccccatgtggttctaaacaaa attcctatagtaaataagtccataagagggtaaggctatctcattttgatttctttttaa ttacaaaaggagcagggcttagagactagatgttagctcaggtggcagagtgccaagcat gcgtgaaggccagtttggatgctagcacctagaagccaagcatgatggcacatgcctacc agcctagtccctggcaggaacgaaggaatgaaggaacaaagggaaggaaggaagaaagga aggaaggaaataaaaataaagtactacttctgctatcatattcccaacaactaaatgctt tctaagaaaaatgtctttctcactgaatccttgccagtactggatattatcattttagcc cagtctttggctctggtatagttgtatgattatgactattattaggactgaaaattaata atatttataaactattctcatcaatagTTTCCAAAAGAAAGACTCATACAACGTGCCCTG S K R K T H T T C P D ACTGCCCTAGCCCCATTGACTTGTCAAACCCCAGTGCTCTGGAAGCTGCCACGGAGTCGC C P S P I D L S N P S A L E A A T E S L TTGCAAAGTTCAACAGTAAGAGCCCCTCAAAAAAGTATGAACTCGTCAAAGTCACCAAGG A K F N S K S P S K K Y E L V K V T K A CTATGAACCAGgtaaagctaaaatgcccatgtcagttacacagtcgtcactgcagggtct M N Q tagcctggttgggggtagggggtggggagggggactgtccaggcgttctgtctgcctaga acctgcgatcctttcctatatctcaattaatttctaggacagccactgtgcattcagtca tattacagctcttgctattccttggaatttgtatgaatagcaggtgacttactttctctc tctctctctctctctctctctctctctctctctctctcgacggggtttctctgtatagcc ctggctgtcctagaactcactctgtagaccaggttggcctcgaactcagaaatctgcctg cctctgcctcccgagtgctgggattaaaggcgtgtgccaccactgcccggcctatgactt gactttctttttttttttttttttttatttttatttattttttttttaaagatttattta ttgattatatgtaagtacactgtagctgtcttcagacactccagaagagggagtcagatc tcattacggatggttgtgagccaccatgtggttgctgggatttgaactctggaccttcgg aagagcagtcgggcgctcctacccactgagccatctcaccagcccatgacttgactttct taaagcaagaagattcttagttttatgtgagcttgggatgaaataggttcagcatttttc tggctgattatggaccctttagcaagacaacttttgcttctttaaggcctggggtaggtg ttgagctttataggttgtggaataagagattcatttatatttcaattgtgtggatagctc ttgcatataaataaaactaaaagcaaagctttatttaaaaaaataagcaaaccaaggcac cgatttttatgggttttttcttttaatttcatttcatgtgtgttggtattttgtctgtgt gagggtattagatcccccttgaactagagttatggacaattgtgggtgccaggaattgaa ctcaggtcctctggaaaaacagtcagtgctcttaaccactgagccatctctccagtcctg attcatattttctaaatagcagaattactgacttggtcttgtgcctgtggggacttcagg 122 APPENDIX

aacagtgaacttgggtgttgacagtgcctcagaatcagttctatacaaggactgttgtca

atcctgagtttcccctggggtaagattttaatgtcttaactcttatttcatggcagTGGG W V TGTCTGGCCCTGCTTACTATGTGGAATATTTGATCAAAGAGGCACCATGTACCAAATCCC S G P A Y Y V E Y L I K E A P C T K S Q AGGCCAGCTGTTCGCTCCAGCACTCTGACTCTGAGtgagtgttcccgactgactgtaca A S C S L Q H S D S E P atctgtatgtgttcacaggtcatcagtacttgtggagcacaaattcccctggggggtgtc acaagtcatcccatgcttccctcccctccccaagcagttagaaactgggcagctcttgat gcagtgataggatttatttgattctaagtgtggtttttatgtaatataattgcaaaaaaa aaatcatgagaggacttggaagacagcctggtgttccattctttgggccactcttcacaa tgtggctaagaaaagtaagggatgagcaccccagcactatcagaaagatactgggaatgg aatcagcgtggtgtgtttgagcaccactgggcaacggtacttttccttgtttatgtatag tggggttttactgagctacagtttacatacaataagatttatcatcttaaagtagataat tccatttttgttatatattcacagacctgagtcaccataaccacctcatatttttaagaa aaaaatacttttattagctctcagaagaaaccccatctctattaagttaacccccgttgc tgccctaagaagaccctagaaactatgaatctgatttctgtcttaatagctttgtctatt ttaacacttcatacaaatgggatcatgcagaggattggggtggggagagatatttatttg aataagaaacttgaccgatgaactccatcagactagccaatgaatgagatcggtgaagtg cagcccccaaagactggccctctgtgctccactccatgccagcacaagaaggtctgcgag atttgatagctcatgaagttctacctagcatggaatatccacctgttcttgccacaggtg catgcagtttctctagaaggttctctgagagcctctgagccatggcattaaagaggaaga agggaagtcagggccaccattttacactaatggattttcccacctggagaaatttctggt cttcatgtgaccaaggaaacatatagaagataggctctgtttgaggcatgcagtttcaga gggttagcatccatggccatcatggtgcatagcatggtagcaggcaagaaggcatggtac tgcagcagtggctggcagcttcatcctgatccacaagtaggaagcagagggagctaaatg ggttcttaaaacctcaaatcccaatcccagtgacacatcccctccaaaaaggccccacct cctaatccttcccaaccagttctaccaaccagggaccaagcattcaaatatatgaggcta tagagccactctcattcaaaccaccacatatggattcactgtaaagaagaaacttagaga tatatgaaaggaaagtttcaatgttttataaaaaaaatagattcaatctagttgtttgac aagagactttattaaatacagaggagtactaggtagccattataaagataataattgaag aaaaaagactatcaaatgtcatacaaaagtttttacaatatattgttcagaaacaaaata gtttttaaaaatatcatatgcaatatgatcatagagtttaaaagaaaaatatacgtgcat agggaaaagattagaagaaaaggaccaaaagattacaggtagtgagagccatgcatggga ttacaggtcatggtcctttcttttgtttgtatagttctaaatagtaatttattacattta taataaaacccctacaatttcattttcaaatagacagaaggttaagggactgagagtgtt tttgctagagaagacaaaccctcattcagaatacattaagggctcctgtagagaagaaaa caccaagcttctaattggccaagggcagacctagggccaggcagtggacagtatgtagac acagggaaaccttcctaaaaacctcagttcccattgctagaatcgagctgcactcaattg tctttagataattcacagggacctggctgggagcccgccattttccaaggcaggagcaga aaaccatggcacagctatgccttacgtaagcggaaagggaggggcttgcatctgcgctca cctgtaacttaatgcccacatgatgcctttccttcttttagCCCGTTGGTATTTGCCAAG V G I C Q G GTTCTACGGTCCAAAGTTCTCTGAGGCACGTTCCTCTGATCCAACCCGTAGAAAAGTCTG S T V Q S S L R H V P L I Q P V E K S V TCACTGTGACTTGTGAGTTCTTCGAATCTCAGgtatttatctttttaaaaacaatgttat V T C E F F E S Q A Q tgtgtttttgcacaacgtaagcgtattgagcacatgtaacatggcacacatgtggagtca gaggataacatggtgtagtttgttctttccttccacctttacatggggtctggggagttg aactcgggtcgctgggcttgcaagagcatgatgttccatgctgagccctaccattctaac cgcccccactctgtgtttttcaatgaaatcaggtgatgtgtttctaggtgcctatttccc aacctcttctggcttcccccaagattcagaatcactttacttaggctctgtaaacccaca catataggagatgagcgtaccctactgttttaggtccagtggcagcaggtgcagaataag ggaagaacactcaggaacaacacagtgaactacacacttgcatagcctaagccttcaagg tgctaaatccagtgtctccaagaagcctctctcacctcagagagaatcacttccgtgaac atgttcattctctctccacagacactaagttcttcctgagacgtagcctgatctagtcag acaggctgcctgggcttcctccctgctctgtccctataagctctgctgccaccccttacc tgtctgggacttggtaccctcatcatctgcctgttgtgatatctaagaccagtgagaagg tttggatgcagggcactatactgggtgtggcatgaaacaaacagtaaaagaggccacctg ctgccactcctgggagcaaaggcaactccctggggaccctgagtacatcctttggcccct ctgaaccactgctctgaagtgaacggcctcatgtccagcatctgcaggtcttagttctag ctgtcccttacgtgttttgttgttgatctttctgtcaagaggcctgttcctgagctcagt FETUB GENOMIC SEQUENCE 123

ggttctcaactgcaggtggcaacacaggggtcacctaagaccattggaaaacacagatgt ttacgttatgattcataagaaaagcgaggttatgaaaataatttcaccacaacatgaact gtctcaaagggtcatggcattaggaaggttgagaaccactggtgtatctggcttctctaa gcattcagaaatgctccaagcagagacaagtgtgttagcccaggcctggaaacccagcac tcaggagatgaggttggaaatgatgaatctgagcctgggctagcttaagaccatgtaaca aacatgcagtcacaaaacagctttgcctggtgtgggttatggcagggcattccctgtaga cttctgcatctaacaacattgttatttcagGCTCAGGTCCCTGGAGATGAGAACCCTGCT V P G D E N P A V T GTTACCCAGGGCCCTCAGAAACTCCCTCAGAAAAACACGGCCCCTACCAGCTCCCCCTCC Q G P Q K L P Q K N T A P T S S P S V T GTAACTGCACCAAGAGGATCCATCCAGCACCTCCCCGAACTGGATGATGAGAAGCCCGAG A P R G S I Q H L P E L D D E K P E E GAGTCCAAGGGAGGGAGCCCTGAGGAAGCCTTTCCTGTGCAGCTGGATCTAACCACCAAT S K G G S P E E A F P V Q L D L T T N P CCCCAGGGGGACACACTAGATGTCTCCTTCCTGTACCTGGAGCCTGGGGACAAGAAGCTG Q G D T L D V S F L Y L E P G D K K L V

GTGGTGCTGCCTTTCCCTGGAAAGGAACAGCGCTCCGCCGAGTGCCCAGGGCCCGAGAAG V L P F P G K E Q R S A E C P G P E K E GAGAACAACCCTCTGGTTCTCCCACCCTGAGACTCCCTAGCAGGGTTTCATAGGGCTATG N N P L V L P P GTCCCCAGCACTAAATGGGAGGTGGTGGGGATTGGGAAGGACACAGACAATGAAATGTAG ACAGGCTAAtaaagtgtgtccttttgatgcttcttggcttcaacctgttgactgaaactg gaaatgagactaaatgcccatgctggagcac

Figure 4.3: Genomic sequence of Fetub. Green letters indicate the cloned mouse promoter sequence MP2 as described in section 3.4.1; red letters indi- cate the cloned mouse promoter sequence MP1 as described in section 3.4.1; bold black letters symbolize the cloned sequences for homologue recombina- tion as described in section 3.1.1 ; capital letters indicate amino acids and underlined capital letters symbolize exons.

Abbreviations

+/+ wildtyp (genotype) +/- heterozygote -/- homozygous knockout Ahsg genetic symbol of fetuin-A BAC bacterial artificial chromosome cDNA copy DNA COC cumulus-oocyte complex DNA desoxyribonucleic acide dpc day post coitus E. coli Escherichia coli ELISA enzyme-linked immunosorbent assay ES cells embryonic stem cells Fx x. inbred generation FCS fetal calf serum FetuB genetic symbol of fetuin-B FSH follicle stimulating hormone HRG histidine-rich glycoprotein kb kilo bases kDa kilo Dalton KO knockout LH luteinizing hormone LPS lipopolysaccharid M molar MCS multiple cloning site MLEC mink lung epithelial cells NC negative control Nx x. backcrossing generation

125 126 ABBREVIATIONS

ON over night ORF open reading frame PAGE polyacrylamide gel electrophoresis PBS phosphate buffered saline PC positive control PCR polymerase chain reaction RPM rounds per minute RT room temperature SDS sodium dodecyl sulfate SPF specific pathogen free TGF transforming growth factor Tris tris-(hydroxymethyl)-aminomethan UTR untranslated region WT wildtype ZP zona pellucida List of Figures

1.1 Fetuin sequence comparison ...... 8 1.2 TGF-β receptor type II homology ...... 9 1.3 Genomic context of the fetuin genes ...... 11 1.4 Transcripts and products of the fetuin genes ...... 11 1.5 An oocyte ...... 15

3.1 The fetuin-B gene targeting vector ...... 48 3.2 The knockout control vector ...... 49 3.3 Scheme of fetuin-B knockout screening strategy ...... 50 3.4 Establishing the knockout screening PCR ...... 51 3.5 Scheme of southern blot screening strategy ...... 52 3.6 Establishing the Southern blot screening ...... 53 3.7 Screening PCR gel electrophoresis ...... 54 3.8 Verifying homologously recombined ES cells by Southern Blot 55 3.9 Fetuin-B genotype analysis ...... 57 3.10 Fetuin-B knockout analysis on protein level ...... 58 3.11 Segregation analysis ...... 59 3.12 Mating calendar ...... 62 3.13 Oocytes’ diameter and zona pellucida thickness ...... 63 3.14 Micrographs of in Vitro cultivated oocytes ...... 64 3.15 Kaplan-Meier developmental curve of oocytes ...... 65 3.16 Micrographs of parthenogenetically developed oocytes . . . . . 66 3.17 X-rays of 12 months old mice ...... 68 3.18 Micrographs of reproductive organs ...... 70 3.19 Micrographs of intestines ...... 71 3.20 Micrographs of liver, kidney, pancreas and spleen ...... 72 3.21 Micrographs of ear and lung ...... 73 3.22 Body weight ...... 75

127 128 LIST OF FIGURES

3.23 Blood pressure and pulse rate graph ...... 76 3.24 Glucose tolerance test ...... 77 3.25 Clinical chemistry screening ...... 79 3.26 Blood coagulation parameter ...... 80 3.27 Sequence of cloning the fetuin-B fusion protein ...... 82 3.28 Fetuin-B cDNA amplification primers and their binding sites on the fetuin-B cDNA ...... 83 3.29 Consensus sequence of sequencing data ...... 85 3.30 Fetuin-B Western blot of transfected COS-7 cells ...... 86 3.31 Purification chromatogram ...... 87 3.32 Analysis of purification products ...... 88 3.33 Precipitation inhibition assay ...... 91 3.34 TGF-β reporter activity in mink lung cells ...... 92 3.35 Fetuin-B serum concentration ...... 94 3.36 Localization of predicted FetuB promoter ...... 95 3.37 Fetuin-B promoter constructs ...... 96

4.1 DBA/2 backcrossing pedigree ...... 115 4.2 C57BL/6 back crossing pedigree ...... 116 4.3 Fetub genomic sequence ...... 123 List of Tables

1.1 Vital statistics on Mus musculus ...... 18

2.1 PCR Master Mix for amplifying the homologous sequences . . 23 2.2 PCR program to amplify the homologous sequences...... 24 2.3 Screening PCR master mix...... 29 2.4 Screening PCR program...... 29 2.5 Genotyping PCR master mix...... 32 2.6 Genotyping PCR program...... 32 2.7 Antibodies used for Western bolt analysis ...... 33 2.8 PCR reaction mix for amplifying Murine fetuin-B cDNA. . . 39 2.9 PCR program for amplifying murine fetuin-B cDNA ...... 39 2.10 Promoter sequences PCR master mix...... 45 2.11 PCR program for amplifying promoter sequences...... 45

3.1 Blastocyst injection and outcome ...... 56 3.2 Mating table 1 ...... 60 3.3 Mating table 2 ...... 61

129

Bibliography

[1] W. M. Brown, N. R. Saunders, K. Mollgard, and K. M. Dziegielewska. Fetuin. An old friend revisited. BioEssays, (14):749–755, 1992.

[2] K. M. Dziegielewska and W. M. Brown. Fetuin. Springer, New York, 1995.

[3] P. Auberger, L. Falquerho, J. O. Contreres, G. L. E. Pages, G. Le Cam, B. L. E. Rossi, and A. Le Cam. Characterization of a natural inhibitor of the insulin receptor tyrosine kinase: cDNA cloning, purification, and anti-mitogenic activity. Cell, (58):631–640, 1989.

[4] G. Rauth, O. Poeschke, E. Fink, M. Eulitz, S. Tippmer, M. Kellerer, H. U. Haering, P. Nawratil, M. Haasemann, W. Jahnen-Dechen, and W. Mueller-Esterl. The nucleotide and partial amino acid sequences of rat fetuin. Identity with the natural tyrosine kinase inhibitor of the rat insulin receptor. Eur. J. Biochem., (204):523–529, 1992.

[5] K. Goto, K. Yoshida, Y. Suzuki, K. Yamamoto, and H. Sinohara. Molecular cloning and sequencing of cDNA encoding plasma coun- tertrypin, a member of mammalian fetuin family, from the mongolian gerbil, Meriones unguiculatus. J. Biochem., (121):619–625, 1997.

[6] N. D. Rawling and A. J. Barrett. Evolution of proteins of the cystatin superfamily. J. Mol. Evol., (30):60–71, 1990.

[7] W. M. Brown and K. M. Dziegielewska. Friends and relations of the cystatin superfamily–new members and their evolution. Protein Sci, 6(1):5–12, 1997.

[8] E. Olivier, E. Soury, J. L. Risler, F. Smih, K. Schneider, K. Lochner, J. Y. Jouzeau, G. H. Fey, and J. P. Salier. A novel set of hepatic

131 132 BIBLIOGRAPHY

mRNAs preferentially expressed during an acute inflammation in rat represents mostly intracellular proteins. Genomics, 57(3):352–64, 1999.

[9] E. Olivier, E. Soury, P. Ruminy, A. Husson, F. Parmentier, M. Daveau, and J. P. Salier. Fetuin-B, a second member of the fetuin family in mammals. Biochem J, 350 Pt 2:589–97, 2000.

[10] J. Kellermann, H. Haupt, E. A. Auerswald, and W. Mueller-Esterl. The arrangement of disulfide loops in human alpha2-HS glycoprotein. similarity to the disulfide bridge structures of cystatins and kininogens. J. Biol. Chem., 264:14121–14128, 1989.

[11] A. Heiss, A. DuChesne, B. Denecke, J. Grotzinger, K. Yamamoto, T. Renne, and W. Jahnen-Dechent. Structural basis of calcification inhibition by alpha 2-HS glycoprotein/fetuin-A. formation of colloidal calciprotein particles. J Biol Chem, 278(15):13333–41, 2003.

[12] M. Demetriou, C. Binkert, B. Sukhu, H. C. Tenenbaum, and J. W. Den- nis. Fetuin/alpha2-HS glycoprotein is a transforming growth factor- beta type II receptor mimic and cytokine antagonist. J Biol Chem, 271(22):12755–61, 1996.

[13] P. Nawratil, S. Lenzen, J. Kellermann, H. Haupt, T. Schinke, W. M¨uller-Esterl,and W. Jahnen-Dechent. Limited proteolysis of hu- man 2-HS glycoprotein/fetuin. J. Biol. Chem., 271:31735–31741, 1996.

[14] D. Kubler, D. Gosenca, M. Wind, H. Heid, I. Friedberg, W. Jahnen- Dechent, and W.D. Lehmann. Proteolytic processing by matrix metal- loproteinases and phosphorylation by protein kinase CK2 of fetuin-A, the major globulin of fetal calf serum. Biochimie, 89(3):410–8, 2007.

[15] B. Denecke, S. Graber, C. Schafer, A. Heiss, M. Woltje, and W. Jahnen- Dechent. Tissue distribution and activity testing suggest a similar but not identical function of fetuin-B and fetuin-A. Biochem J, 376(Pt 1):135–45, 2003.

[16] P. Arnaud, L. Miribel, and D. L. Emerson. Alpha 2-HS glycoprotein. Methods Enzymol, 163:431–41, 1988. BIBLIOGRAPHY 133

[17] M. Woeltje, B. Tschoeke, V. von Bulow, R. Westenfeld, B. Denecke, S. Graber, and W. Jahnen-Dechent. CCAAT enhancer binding pro- tein beta and hepatocyte nuclear factor 3beta are necessary and suf- ficient to mediate dexamethasone-induced up-regulation of alpha2hs- glycoprotein/fetuin-a gene expression. J Mol Endocrinol, 36(2):261–77, 2007.

[18] S. J. Hsu, H. Nagase, and A. Balmain. Identification of fetuin-B as a member of a cystatin-like gene family on mouse chromosome 16 with tumor suppressor activity. Genome, 47(5):931–46, 2004.

[19] K. O. Pedersen. Fetuin, a new globin isolated from serum. Nature, 154:575–570, 1944.

[20] T. Ohnishi, O. Nakamura, N. Arakaki, and Y. Daikuhara. Effect of phosphorylated rat fetuin on the growth of hepatocytes in primary cul- ture in the presence of human hepatocyte-growth factor. Evidence that phosphorylated fetuin is a natural modulator of hepatocyte-growth fac- tor. Eur J Biochem, 243(3):753–61, 1997.

[21] K. M. Dziegielewska and N. A. Andersen. The fetal glycoprotein, fetuin, counteracts ill-effects of the bacterial endotoxin, lipopolysaccharide, in pregnancy. Biol Neonate, 74(5):372–5, 1998.

[22] H. Wang, M. Zhang, K. Soda, A. Sama, and K. J. Tracey. Fetuin protects the fetus from TNF. Lancet, 350(9081):861–2, 1997.

[23] H. Wang, M. Zhang, M. Bianchi, B. Sherry, A. Sama, and K. J. Tracey. Fetuin (alpha2-HS-glycoprotein) opsonizes cationic macrophagedeacti- vating molecules. Proc Natl Acad Sci U S A, 95(24):14429–34, 1998.

[24] C. Schafer, A. Heiss, A. Schwarz, R. Westenfeld, M. Ketteler, J. Floege, W. Muller-Esterl, T. Schinke, and W. Jahnen-Dechent. The serum pro- tein alpha 2-Heremans-Schmid glycoprotein/fetuin-A is a systemically acting inhibitor of ectopic calcification. J Clin Invest, 112(3):357–66, 2003.

[25] W. Jahnen-Dechent, T. Schinke, A. Trindl, W. Muller-Esterl, F. Sablitzky, S. Kaiser, and M. Blessing. Cloning and targeted deletion of the mouse fetuin gene. J Biol Chem, 272(50):31496–503, 1997. 134 BIBLIOGRAPHY

[26] M. W. Merx, C. Schafer, R. Westenfeld, V. Brandenburg, S. Hida- jat, C. Weber, M. Ketteler, and W. Jahnen-Dechent. Myocardial stiffness, cardiac remodeling, and diastolic dysfunction in calcification- prone fetuin-A-deficient mice. J Am Soc Nephrol, 16(11):3357–64, 2005.

[27] R. Westenfeld, W. Jahnen-Dechent, and M. Ketteler. Vascular cal- cification and fetuin-A deficiency in chronic kidney disease. Trends Cardiovasc Med, 17(4):124–8, 2007.

[28] S. T. Mathews, G. P. Singh, M. Ranalletta, V. J. Cintron, X. Qiang, A. S. Goustin, K. L. Jen, M. J. Charron, W. Jahnen-Dechent, and G. Grunberger. Improved insulin sensitivity and resistance to weight gain in mice null for the Ahsg gene. Diabetes, 51(8):2450–8, 2002.

[29] S. T. Mathews, S. Rakhade, X. Zhou, G. C. Parker, D. V. Coscina, and G. Grunberger. Fetuin-null mice are protected against obesity and insulin resistance associated with aging. Biochem Biophys Res Commun, 350(2):437–43, 2006.

[30] M. L. Leite-Browning, L. J. McCawley, W. Jahnen-Dechent, L. E. Jr. King, L. M. Matrisian, and J. Ochieng. Alpha 2-HS glycopro- tein (fetuin-A) modulates murine skin tumorigenesis. Int J Oncol, 25(2):319–24, 2004.

[31] C. J. Swallow, E. A. Partridge, J. C. Macmillan, T. Tajirian, G. M. DiGuglielmo, K. Hay, M. Szweras, W. Jahnen-Dechent, J. L. Wrana, M. Redston, S. Gallinger, and J. W. Dennis. alpha2HS-glycoprotein, an antagonist of transforming growth factor beta in vivo, inhibits in- testinal tumor progression. Cancer Res, 64(18):6402–9, 2004.

[32] M. N. Kundranda, M. Henderson, K. J. Carter, L. Gorden, A. Bin- hazim, S. Ray, T. Baptiste, M. Shokrani, M. L. Leite-Browning, W. Jahnen-Dechent, L. M. Matrisian, and J. Ochieng. The serum gly- coprotein fetuin-A promotes lewis lung carcinoma tumorigenesis via adhesive-dependent and adhesive-independent mechanisms. Cancer Res, 65(2):499–506, 2005. BIBLIOGRAPHY 135

[33] M. E. Pepling and A. C. Spradling. Mouse ovarian germ cell cysts undergo programmed breakdown to form primordial follicles. Dev Biol, 234(2):339–51, 2001.

[34] Y. Masui and H. J. Clarke. Oocyte maturation. Int Rev Cytol., 57:185– 282, 1979.

[35] Z. B. Tong, L. Gold, K. E. Pfeifer, H. Dorward, E. Lee, C. A. Bondy, J. Dean, and L. M. Nelson. Mater, a maternal effect gene required for early embryonic development in mice. Nat Genet, 26(3):267–8, 2000.

[36] X. Wu, M. M. Viveiros, J. J. Eppig, Y. Bai, S. L. Fitzpatrick, and M. M. Matzuk. Zygote arrest 1 (Zar1) is a novel maternal-effect gene critical for the oocyte-to-embryo transition. Nat Genet, 33(2):187–91, 2003.

[37] R. Bachvarova. A comprehensive synthesis. Developmental Biology, 1:453–524, 1985.

[38] P. M. Wassarman, J. D. Bleil, H. M. Florman, J. M. Greve, R. J. Roller, G. S. Salzmann, and F. G. Samuels. The mouse egg’s receptor for sperm: what is it and how does it work? Cold Spring Harb Symp Quant Biol, 50:11–9, 1985.

[39] R. A. Kinloch, S. A. Lira, S. Mortillo, M. Schickler, R. J. Roller, and P. M. Wassarman. Regulation of expression of mZP3, the sperm recep- tor gene, during mouse development. Molecular basis of morphogenesis, pages 19–33, 1993.

[40] R. Yanagimachi. Mammalian fertilization. The Physiology of Repro- duction, 1:189–317, 1994.

[41] P. M. Wassarman. Zona pellucida glycoproteins. Annu Rev Biochem, 57:415–42, 1988.

[42] J. M. Greve and P. M. Wassarman. Mouse egg extracellular coat is a matrix of interconnected filaments possessing a structural repeat. J Mol Biol, 181(2):253–64, 1985. 136 BIBLIOGRAPHY

[43] J. D. Bleil and P.M. Wassarman. Identification of a ZP3-binding pro- tein on acrosome-intact mouse sperm by photoaffinity crosslinking. Proc Natl Acad Sci U S A., 87(14):5563–7, 1990.

[44] A. Nagy, M. Gertsenstein, K. Vintersten, and R. Behringer. Manipu- lating the Mouse Embryo. Cold Spring Harbor Laboratory Press, 2003.

[45] H. M. Florman, K. B. Bechtol, and P. M. Wassarman. Enzymatic dissection of the functions of the mouse egg’s receptor for sperm. Dev Biol, 106(1):243–55, 1984.

[46] P. M. Wassarman. Profile of a mammalian sperm receptor. Develop- ment, 108(1):1–17, 1990.

[47] P. M. Wassarman, L. Jovine, and E. S. Litscher. A profile of fertilization in mammals. Nat Cell Biol, 3(2):59–64, 2001.

[48] P. Primakoff and D. G. Myles. Penetration, adhesion, and fusion in mammalian sperm-egg interaction. Science, 296(5576):2183–5, 2002.

[49] J. P. Evans and H. M. Florman. The state of the union: the cell biology of fertilization. Nat Cell Biol, 4 Suppl:s57–63, 2002.

[50] F. Le Naour, E. Rubinstein, C. Jasmin, M. Prenant, and C. Boucheix. Severely reduced female fertility in CD9-deficient mice. Science, 287(5451):319–21, 2000.

[51] K. Miyado, G. Yamada, S. Yamada, H. Hasuwa, Y. Nakamura, F. Ryu, K. Suzuki, K. Kosai, K. Inoue, A. Ogura, M. Okabe, and E. Mekada. Requirement of CD9 on the egg plasma membrane for fertilization. Science, 287(5451):321–4, 2000.

[52] K. Kaji, S. Oda, T. Shikano, T. Ohnuki, Y. Uematsu, J. Sakagami, N. Tada, S. Miyazaki, and A. Kudo. The gamete fusion process is defective in eggs of Cd9-deficient mice. Nat Genet, 24(3):279–82, 2000.

[53] D. A. Ellerman, C. Ha, P. Primakoff, D. G. Myles, and G. S. Dveksler. Direct binding of the ligand PSG17 to CD9 requires a CD9 site essential for sperm-egg fusion. Mol Biol Cell, 14(12):5098–103, 2003. BIBLIOGRAPHY 137

[54] W. H. Colledge, M.B. Carlton, Udy G. B., and M. J. Evans. Disrup- tion of c-mos causes parthenogenetic development of unfertilized mouse eggs. Nature, 370(6484):65–8, 1994.

[55] N. Hashimoto, N. Watanabe, Y. Furuta, H. Tamemoto, N. Sagata, M. Yokoyama, K. Okazaki, M. Nagayoshi, N. Takeda, and Y. Ikawa. Parthenogenetic activation of oocytes in c-mos-deficient mice. Nature, 370(6484):68–71, 1994.

[56] S. L. Mansour, K. R. Thomas, and M.R. Capecchi. Disruption of the protooncogene int-2 in mouse embryo-derived stem cells: a gen- eral strategy for targeting mutations to non-selectable genes. Nature, 336:348–52, 1988.

[57] G. R. Martin. Isolation of a pluripotent cell line from early mouse embryos cultured in medium conditioned by teratocarcinoma cells. Proc Natl Acad Sci U S A, 78:7634–8, 1981.

[58] M. J. Evans and M. H. Kaufman. Establishment in culture of pluripo- tent cells from mouse embryos. Nature, 292:154–6, 1981.

[59] O. Smithies, R. G. Gregg, S. S. Boggs, M. A. Koralewski, and R. S. Kucherlapati. Insertion of DNA sequences into the human chro- mosomal beta-globin locus by homologous recombination. Nature, 317(6034):230–4, 1985.

[60] Frederick M. Ausubel. Current Protocols in Molecular Biology. 4 vols. John Wiley and Sons Inc, Indianapolis, 1994.

[61] H. Fuchs, K. Schughart, E. Wolf, R. Balling, and M. Hrabe de Angelis. Screening for dysmorphological abnormalities–a powerful tool to isolate new mouse mutants. Mamm Genome, 11(7):528–30, 2000.

[62] R. Weiskirchen, J. Kneifel, S. Weiskirchen, E. van de Leur, D. Kunz, and A. M. Gressner. Comparative evaluation of gene delivery devices in primary cultures of rat hepatic stellate cells and rat myofibroblasts. BMC Cell Biol, 1:4, 2000.

[63] T. Schinke, C. Amendt, A. Trindl, O. Poschke, W. Muller-Esterl, and W. Jahnen-Dechent. The serum protein alpha2-HS glycoprotein/fetuin 138 BIBLIOGRAPHY

inhibits apatite formation in vitro and in mineralizing calvaria cells. a possible role in mineralization and calcium homeostasis. J Biol Chem, 271(34):20789–96, 1996.

[64] M. Abe, J. G. Harpel, C. N. Metz, I. Nunes, D. J. Loskutoff, and D. B. Rifkin. An assay for transforming growth factor-beta using cells transfected with a plasminogen activator inhibitor-1 promoter- luciferase construct. Anal Biochem, 216(2):276–84, 1994.

[65] R. H. Waterston, K. Lindblad-Toh, E. Birney, J. Rogers, J. F. Abril, P. Agarwal, R. Agarwala, R. Ainscough, M. Alexandersson, and P. An. Initial sequencing and comparative analysis of the mouse genome. Na- ture, 420:520–562, 2002.

[66] N. Tsuchida-Straeten, S. Ensslen, C. Schafer, M. Woltje, B. Denecke, M. Moser, S. Graber, S. Wakabayashi, T. Koide, and W. Jahnen- Dechent. Enhanced blood coagulation and fibrinolysis in mice lacking histidine-rich glycoprotein (HRG). J Thromb Haemost, 3(5):865.72, 2005.

[67] P. R. Srinivas, A. S. Wagner, L. V. Reddy, D. D. Deutsch, M. A. Leon, A. S. Goustin, and G. Grunberger. Serum alpha 2-HS-glycoprotein is an inhibitor of the human insulin receptor at the tyrosine kinase level. Mol Endocrinol, 7(11):1445–55, 1993.

[68] S. T. Mathews, N. Chellam, P. R. Srinivas, V. J. Cintron, M. A. Leon, A. S. Goustin, and G. Grunberger. Alpha2-HSG, a specific inhibitor of insulin receptor autophosphorylation, interacts with the insulin recep- tor. Mol Cell Endocrinol, 164(1-2):87–98, 2000.

[69] H. Chen, P. R. Srinivas, L. N. Cong, Y. Li, G. Grunberger, and M. J. Quon. Alpha2-Heremans Schmid glycoprotein inhibits insulin- stimulated Elk-1 phosphorylation, but not glucose transport, in rat adipose cells. Endocrinology, 139(10):4147–54, 1998.

[70] A. Bradley, M. Evans, M. H. Kaufman, and E. Robertson. Formation of germ-line chimaeras from embryo-derived teratocarcinoma cell lines. Nature, 309(5965):255–6, 1984. BIBLIOGRAPHY 139

[71] F. A. Lemckert, J. D. Sedgwick, and H. Korner. Gene targeting in C57BL/6 ES cells. Successful germ line transmission using recipient BALB/c blastocysts developmentally matured in vitro. Nucleic Acids Res, 25(4):917–8, 1997.

[72] K. Eggan, H. Akutsu, J. Loring, L. Jackson-Grusby, M. Klemm, 3rd Rideout, W. M., R. Yanagimachi, and R. Jaenisch. Hybrid vigor, fetal overgrowth, and viability of mice derived by nuclear cloning and tetraploid embryo complementation. Proc Natl Acad Sci U S A, 98(11):6209–14, 2001.

[73] E. S. Litscher and P. M. Wassarman. Egg extracellular coat proteins: from fish to mammals. Histol Histopathol, 22(3):337–47, 2007.

[74] Nimpf J Waclawek M, Foisner R and Schneider WJ. The chicken ho- mologue of zona pellucida protein-3 is synthesized by granulosa cells. Biol Reprod., 59(5):1230–9, 1998.

[75] H. Sato, S. Kajikawa, S. Kuroda, Y. Horisawa, N. Nakamura, N. Kaga, C. Kakinuma, K. Kato, H. Morishita, H. Niwa, and J. Miyazaki. Impaired fertility in female mice lacking urinary trypsin inhibitor. Biochem Biophys Res Commun, 281(5):1154–60, 2001.

[76] L. Zhuo, M. Yoneda, M. Zhao, W. Yingsung, N. Yoshida, Y. Kitagawa, K. Kawamura, T. Suzuki, and K. Kimata. Defect in SHAP-hyaluronan complex causes severe female infertility. A study by inactivation of the bikunin gene in mice. J Biol Chem, 276(11):7693–6, 2001.

[77] M. Ruggiu, R. Speed, M. Taggart, S. J. McKay, F. Kilanowski, P. Saun- ders, J. Dorin, and H. J. Cooke. The mouse Dazla gene encodes a cy- toplasmic protein essential for gametogenesis. Nature, 389(6646):73–7, 1997.

[78] S. M. Soyal, A. Amleh, and J. Dean. FIGalpha, a germ cell-specific transcription factor required for ovarian follicle formation. Develop- ment, 127(21):4645–54, 2000.

[79] J. Dong, D. F. Albertini, K. Nishimori, T. R. Kumar, N. Lu, and M. M. Matzuk. Growth differentiation factor-9 is required during early ovarian folliculogenesis. Nature, 383(6600):531–5, 1996. 140 BIBLIOGRAPHY

[80] T. R. Kumar, Y. Wang, N. Lu, and M. M. Matzuk. Follicle stimulat- ing hormone is required for ovarian follicle maturation but not male fertility. Nat Genet, 15(2):201–4, 1997.

[81] J. E. Dinchuk, B. D. Car, R. J. Focht, J. J. Johnston, B. D. Jaffee, M. B. Covington, N. R. Contel, V. M. Eng, R. J. Collins, and P. M. Czerniak. Renal abnormalities and an altered inflammatory response in mice lacking cyclooxygenase II. Nature, 378(6555):406–9, 1995.

[82] J. P. Lydon, F. J. DeMayo, O. M. Conneely, and B. W. O’Malley. Re- productive phenotpes of the progesterone receptor null mutant mouse. J Steroid Biochem Mol Biol, 56(1-6):67–77, 1996.

[83] C. L. Stewart. Leukaemia inhibitory factor and the regulation of pre- implantation development of the mammalian embryo. Mol Reprod Dev, 39(2):233–8, 1994.

[84] Y. Choi, Y. Qin, M. F. Berger, D. J. Ballow, M. L. Bulyk, and A. Ra- jkovic. Microarray analyses of newborn mouse ovaries lacking Nobox. Biol Reprod, 2007.

[85] S. B. Ramos, D. J. Stumpo, E. A. Kennington, R. S. Phillips, C. B. Bock, F. Ribeiro-Neto, and P. J. Blackshear. The CCCH tandem zinc- finger protein Zfp36l2 is crucial for female fertility and early embryonic development. Development, 131(19):4883–93, 2004.

[86] A. L. Staros and G. J. Killian. In vitro association of six oviductal fluid proteins with the bovine zona pellucida. J Reprod Fertil, 112(1):131–7, 1998.

[87] N. S. Martus, H. G. Verhage, P. A. Mavrogianis, and J. K. Thi- bodeaux. Enhancement of bovine oocyte fertilization in vitro with a bovine oviductal specific glycoprotein. J Reprod Fertil, 113(2):323–9, 1998.

[88] H. Abe and H. Hoshi. Bovine oviductal epithelial cells: their cell culture and applications in studies for reproductive biology. Cytotechnology, 23:171–183, 1997. BIBLIOGRAPHY 141

[89] D. E. Boatman and G. E. Magnoni. Identification of a sperm pen- etration factor in the oviduct of the golden hamster. Biol Reprod, 52(1):199–207, 1995.

[90] R. H. F. Hunter. The Fallopian Tubes: Their Role in Fertilty and Infertility. Springer-Verlag, Berlin, 1988.

[91] Y. Araki, M. Nohara, H. Yoshida-Komiya, and T. Kuramochi. Effect of a null mutation of the oviduct-specific glycoprotein gene on mouse fertilization. Biochem J, 374:551–557, 2003.

[92] T. Rankin, M. Familari, E. Lee, A. Ginsberg, N. Dwyer, J. Blanchette- Mackie, J. Drago, H. Westphal, and J. Dean. Mice homozygous for an insertional mutation in the Zp3 gene lack a zona pellucida and are infertile. Development, 122(9):2903–10, 1996.

[93] P. M. Wassarman, H. Qi, and E. S. Litscher. Mutant female mice carrying a single mZP3 allele produce eggs with a thin zona pellucida, but reproduce normally. Proc Biol Sci, 264(1380):323–8, 1997.

[94] A. C. Schroeder, R. M. Schultz, G. S. Kopf, F. R. Taylor, R. B. Becker, and J. J. Eppig. Fetuin inhibits zona pellucida hardening and conver- sion of ZP2 to ZP2f during spontaneous mouse oocyte maturation in vitro in the absence of serum. Biol Reprod, 43(5):891–7, 1990.

[95] P. Kalab, G. S. Kopf, and R. M. Schultz. Modifications of the mouse zona pellucida during oocyte maturation and egg activation: effects of newborn calf serum and fetuin. Biol Reprod, 45(5):783–7, 1991.

[96] P. Kalab, R. M. Schultz, and G. S. Kopf. Modifications of the mouse zona pellucida during oocyte maturation: inhibitory effects of follicular fluid, fetuin, and alpha 2HS-glycoprotein. Biol Reprod, 49(3):561–7, 1993.

[97] N. Motosugi, J. E. Dietrich, Z. Polanski, D. Solter, and T. Hiiragi. Space asymmetry directs preferential sperm entry in the absence of polarity in the mouse oocyte. PLoS Biol, 4(5):Epub135, 2006.

[98] S. A. Coonrod, S. Naaby-Hansen, J. Shetty, H. Shibahara, M. Chen, J. M. White, and J. C. Herr. Treatment of mouse oocytes with PI-PLC 142 BIBLIOGRAPHY

releases 70-kDa (pI 5) and 35- to 45-kDa (pI 5.5) protein clusters from the egg surface and inhibits sperm-oolemma binding and fusion. Dev Biol, 207(2):334–49, 1999.

[99] J. A. Alfieri, A. D. Martin, J. Takeda, G. Kondoh, D. G. Myles, and P. Primakoff. Infertility in female mice with an oocyte-specific knockout of GPI-anchored proteins. J Cell Sci, 116(Pt 11):2149–55, 2003.

[100] S. Ikeyama, M. Koyama, M. Yamaoko, R. Sasada, and M. Miyake. Suppression of cell motility and metastasis by transfection with human motility-related protein (MRP-1/CD9) DNA. J Exp Med, 177(5):1231– 7, 1993.

[101] H. Fuchs, T. Lisse, K. Abe, and M. Hrab´ede Angelis. Standards of Mouse Model Phenotyping. WILEY-VCH, Weinheim, 2006.

[102] W. F. Loeb and F. W. Quimby. The Clinical Chemistry of Laboratory Animals. Taylor and Francis, Philadelphia, 1999. Acknowledgements

It is a challenge to complete a thesis; however, thanking everyone who con- tributed to it is an even greater one. So many people were involved either in a professional or in a private way and I would like to thank all of them! In particular I would like to take this opportunity to express my gratitude to Professor Willi Jahnen-Dechent for providing the challenging topic and for his support during my thesis. Additionally I would like to thank Professor Lothar Elling for his interest and for co-refereeing my thesis. I would like to thank Dr. Bernd Denecke for assisting me in the lab, as well as Dr. Cora Sch¨aferand Dr. Silke Ensslen for guiding my mouse work. Furthermore, I would like to thank Professor Hubert Schorle and his team for performing the ES-cell transfection and the blastocyst injection, as well as PD Ralf Weiskirchen and his team for the adenovirus production. I would like to thank Dr. Thomas Ren´efor performing the serum-chemistry screening. This work was supported by the German Research Foundation (Deutsche Forschungsgemeinschaft) under Graduate School Grant GRK 1035 Biointer- face where I met my husband. Special thanks for that. Finally, I would like to thank my family and friends for their understanding and support during my studies.

143

Curriculum Vitae

Jennifer Denise Wessling geb. Goldstein, 3. M¨arz1978 in Aachen

Schule & Studium 1984 – 1988 Besuch der Grundschule Am H¨oflingin Aachen

1988 – 1997 Einhard Gymnasium in Aachen, Abschluss Abitur

1997 – 2003 Studium an der RWTH Aachen, Studiengang Biologie mit Hauptfach Molekularbiologie (Abschluss Diplom)

2002 – 2003 Diplomarbeit am Institut f¨urPathologie, Universit¨ats- klinikum Bonn Promotion 2003 – 2004 Wissenschaftliche Angestellte am Institut f¨urBiomedi- zinische Technologien - Zell- und Molekularbiologie an Grenzfl¨achen der RWTH Aachen

2004 – 2007 Stipendiatin des Graduiertenkollegs 1035 (Biointerface) der deutschen Forschungsgemeinschaft, t¨atigam Institut f¨urBiomedizinische Technologien - Zell- und Molekular- biologie an Grenzfl¨achen der RWTH Aachen