Comprehensive Summaries of Uppsala Dissertations from the Faculty of Medicine 1394

Hormonal Regulation of Neural Stem Cell Proliferation and Fate Determination

BY KARIN BRÄNNVALL

ACTA UNIVERSITATIS UPSALIENSIS UPPSALA 2004                             !!" !#$% &   ' &    & (    )*   &  +,      -      . ' ,

  /   0, !!", 1   2'  & 3  4   ( &   *     , 5       ,                 6#", 6 ,    , 743 #8%%"8 8%

4        &   &  -   &         , 7   '         )34+  &          -   & ' &    &        &      & -, 5       '  &      )34+  &   &            ,      &   -       '  34  &   &         , 7       &  &       '        '     -       98      '   )9841+ - :     , 5          '   )258+ -        -   '     ' 34, ; &  '  -   '        &&  34<    34  &       ' , .  ' =     &  -       &     &     > , 7      '   -       &       ' &&   '  , 5   ?      ?    6!@  34  &      '    '  &   &&  34  ,       9841     '     '  &           <   34  &  -   , *  258    -  34  &         - '   &&       ' 258       34  >, 7        '     -  &  34 '  -     &      &   ' , 5  -   &    9841  34  '   258   34  >,

   3     ( &   *      1   5    '    

  !     "    ! " # $! % &'(!      ! )*(&+,-  !  

A 0  /   !!"

7443 !B8C"C 743 #8%%"8 8%  $  $$$ 8" #" ) $DD ,>,D E F $  $$$ 8" #"+ List of Papers

I Brannvall, K., Korhonen, L., Lindholm, D. (2002). Estrogen- -dependent regulation of neural stem cell proliferation and differentiation. Mol Cell. Neurosci. 21(3):512-20. II Brännvall, K.,* Korhonen, L.,* Skoglösa, Y., Lindholm, D. (2003). Tumor suppressor gene BRCA-1 is expressed by embryonic and adult neural stem cells and involved in cell proliferation J. Neurosci Res. 71(6):769-76. (*equal contribution). III Brännvall, K., Bogdanovic, N., Lindholm, D. 19-Nortestosterone Influences Neural Stem Cell Proliferation and Neurogenesis in the Rat Brain. Submitted to EJN IV Brännvall, K., Ingvarsson, H., Lindholm, D. D-MSH Dependent Regulation of Neural Stem Cell Proliferation. Manuscript.

Reprints were made by permission from the publishers. Pictures on the cover:

Embryonic neurospheres stained for:

E-III-tubulin on the front page Nestin on the pack page

All figures appearing in this thesis are made by Karin Brännvall Table of Contents

INTRODUCTION ...... 9 Stem Cells ...... 9 Totipotent, Multipotent and Pluripotent Stem Cells...... 10 DifferentiatioDifferentiationn Potential of Somatic Stem Cells...... 11 Development of the Nervous System...... 12 Formation of Brain and Spinal Cord ...... 12 Neural Stem Cells...... 13 The life of a Neural Stem Cell ...... 14 The Cell Cycle...... 15 Notch and Numb Pathway...... 16 Cell Fate Specific Transcription Factors: the bHLH Proteins...... 17 Differentiation and Fate Determination...... 18 Mature Cell Types within the Central Nervous System...... 21 Hormones...... 24 Sex Hormones...... 24 17E-Estradiol...... 25 19-Nortestosterone ...... 28 The Melanocortin System ...... 29 Melanocortins in the Brain...... 30 BRCA-1 ...... 30 BRCA-1 Structure...... 31 BRCA-1 Expression...... 31 BRCA-1 Mutant Mice...... 32 Estrogen Dependent Regulation of BRCA-1 ...... 32

MATERIALS AND METHODS...... 33 Experimental Animals...... 33 Nandrolone/BrdU Injections ...... 33 Cell Culture...... 33 Primary Neural Stem Cells...... 33 Cell Biology ...... 34 Western blotting ...... 34 Cell Proliferation...... 34 Cell Differentiation ...... 35 Immunocytochemistry...... 35 Immunohistochemistry...... 36 Stereology ...... 37 Molecular Biology ...... 39 RT-PCR...... 39

AIMS OF THE PRESENT INVESTIGATION...... 40 RESULTS AND DISCUSSION ...... 41 Estrogen Affects the Proliferation and Fate Determination of NSCs (PaperI) ...... 41 BRCA-1 is localized to Proliferating Neural Precursors and Down Regulated upon Differentiation (Paper II)...... 41 19-Nortestosterone Influences Neural Stem Cell Proliferation and Neurogenesis in the Rat Brain (PaperIII)...... 42 D-MSH is a Mitogen for Embryonic NSCs (Paper IV)...... 42 GENERAL DISCUSSION AND FUTURE PERSPECTIVE ...... 44 ACKNOWLEDGEMENT ...... 47 REFERENCES ...... 49 ABBREVIATIONS

AAS Anabolic Androgenic Steroids AF Activation Function D-MSH D-Melanocyte Stimulating Hormone apoE apolipoprotein E AR/ARs /Androgen Receptors ARKO Androgen Receptor Knock Out ArKO Aromatase Knock Out BERKO E Knock Out bFGF basic Fibroblast Growth Factor bHLH basic Helix Loop Helix BMP/BMPs Bone Morphogenetic Protein/s BRCA Breast Cancer Susceptibility Gene BRCT BRCA-1 C-terminal repeat cAMP cyclic Adenosine Mono Phosphate cdc cell division control cdk cyclin dependent kinase CNP-ase 2´-3´-Cyclic Nucleotide-3´-Phosphodiesterase CNS Central Nervous System CNTF Ciliary Neurotrophic Factor DG Dentate gyrus DHT Dihydrotestosterone E Embryonic day EGF Epidermal Growth Factor ER/ERs Estrogen Receptor/Estrogen Receptors erk extra-cellular signal-related kinase ERKO Estrogen Receptor D Knock Out ES Cell Embryonic Stem Cell GFAP Glial Fibrillary Acidic Protein Hes Hairy and enhancer of split homolog HRT Hormone Replacement Therapy ICN Intra Cellular Notch Id Inhibitor of Differentiation LIF Leukemia Inhibitory Factor MAP kinase Mitogen activated kinase MCR Melanocortin receptor Ngn Neurogenin NR/NRs /Nuclear Receptors NSC/NSCs Neural Stem Cell/Neural Stem Cells OB Olfactory Bulb P Post natal day PDGF Platelet Derived Growth Factor PI-3 kinase Phosphatidyl Inositol-3 kinase pRb RA Retinoic Acid RMS Rostral Migratory Stream Shh Sonic hedge hog SVZ Sub Ventricular Zone T3 tri-iodo-Thyronine INTRODUCTION

Considering the difference in morphology and function of the cell types present in an adult organism, it is fascinating that they all have both the same genetic setup and also arise from one single cell, the fertilized egg: the first stem cell. This stem cell contains not only the blue print, but also the potential to form the entire offspring. Initially, the fertilized egg divides symmetrically, giving rise to daughter cells with interchangeable fate. Later during development, stem cells start to divide asymmetrically, giving rise to daughter cells with different fates. In the adult, only a small fraction of the total cell number will have maintained their stem cell potential. Since cells in organs such as the bone marrow are constantly replaced; cells with stem cell potential must reside in these tissues, even in the adult. Sur- prisingly, stem cells are also found in for instance the adult human brain, an organ made up of cells which are not typically considered proliferative. The discovery of stem cells in the adult human brain has changed the classi- cal view of the fully developed brain as incapable of forming new neurons. In fact, neurogenesis is on going in the dentate gyrus (DG), a region in hippocam- pus involved in formation of memory. At the present moment, the drugs used for treating patients with Alzheimer’s and Parkinson’s disease only slow down the neurodegeneration. In the future, the possibility to either transplant stem cells into the brain or administer drugs which affect the NSCs in situ might offer an alternative to today’s treatment strategy. In this introduction I will discuss some of the properties which define stem cells in general. Before discussing NSCs in particular, a background to the de- velopment of the nervous system is out-lined. Then I will discuss the three major systems involved in regulation of NSC proliferation and fate determination. Finally, the hormones estrogen, nandrolone and D-melanocyte stimulating hor- mone (D-MSH), as well as the tumor suppressor BRCA-1 will be introduced with emphasis on actions in the brain.

Stem Cells Stem cells are cells which can go through both symmetric and asymmetric cell division, are self renewing and have a high degree of potency (Anderson, 2001). The self renewal capacity results in that at least one daughter cell will have the

9 same potency as the mother cell (Fig1A, B). The high degree of potency results in the ability to form a large number of different mature cell types which will be discussed below.

A B SC Figure 1. Symmetric (A) versus SC asymmetric (B) cell division. A mother cell is forming daughter cells with interchangeable fate (A) and different fates (B). SC, SC SC SC P stem cell; P, Precursor cell.

Totipotent, Multipotent and Pluripotent Stem Cells The ultimate stem cell is the fertilized egg. This cell is completely totipotent, which means that it can form the entire embryo and the extra embryonic tissues such as the placenta. When the fertilized egg has undergone approximately twelve cell divisions, it arrives at a developmental stage called blastula. Inside the inner cell mass of this blastula, the so called embryonic stem (ES) cells reside. These cells are com- pletely undifferentiated, rapidly dividing cells with the potential to form the entire embryo, but not the extra embryonic tissues and are therefore called pluri- potent. Because of its pluripotency, ES cells are used for generation of trans- genic animals. As the embryo develops, stem cells start to undergo asymmetric cell divi- sions, and different cell lineages are formed. These stem cells, which can be isolated from different parts of the developing and adult body, are often referred to as somatic stem cells (from ‘soma’ Greek word for body). In general, somatic stem cells are multipotent, which means that they can form at least two types of differentiated mature progenies (Fig2).

Fertilized Blastula Embryonic Somatic Adult Somatic egg (ES cells) Stem Cells Stem Cells

Figure 2. ESC ASC Different types of stem cells. Totipotent Pluripotent Multipotent Multipotent ESC, embryonic somatic stem cell; ASC, adult somatic stem Embryo & extra Embryo > 2 different > 2 different embryonic tissues mature cell types mature cell types cell.

10 Differentiation Potential of Somatic Stem Cells Transdifferentiation and Cell Fusion In vivo, somatic stem cells are forming only the different progeny found in the tissue of origin. However, recent studies indicate that some stem cell lineages have a higher developmental plasticity, in particular in vitro. For example, pro- genitors from the adult bone marrow have been able to transdifferentiate into brain cells (Kopen et al., 1999; Mezey et al., 2000; Brazelton et al., 2000), mus- cle cells (Ferrari et al., 1998), myocards (Orlic et al., 2001) and hepatocytes (Lagasse et al., 2000). This surprising plasticity is not only confined to bone marrow cells. Adult neural stem cells injected into an early developing embryo, formed both skeletal myotubes (Galli et al., 2000) and cells which arise from all three germ layers (Clarke et al., 2000). In addition, muscle precursors, liver cells and skin cells have also shown to transdifferentiate into adipocytes, pancreatic cells and neurons respectively (Hu et al., 1995; Overturf et al., 1997; Toma et al., 2001). These studies indicate that some somatic stem cell lineages can be dedifferentiated and reprogrammed to form other lineages when subjected to the appropriate local environment. In 2002 Terada and colleagues studied the ploidity of ES cells co-cultured with mouse bone marrow cells. This study revealed that a fraction of the ES cells fused with the bone marrow cells, thereby forming a tetraploid cell contain- ing both the ES cell and the blood specific markers. In addition to bone marrow cells, mouse NSCs have also been shown to fuse with co-cultured ES cells (Ying et al., 2002). While this cell fusion is quite rare, one in 104 to one in 105(Ying et al., 2002; Terada et al., 2002), transdifferentiation is reported to occur at the much higher frequency of 7-57 % (Galli et al., 2000; Rietze et al., 2001). A fraction of stem cells have the ability to fuse with other cells, a capacity which was not anticipated; as a result, in pioneering studies only the fate of the stem cell was tracked using a tag. However, recently a study, in which both the fate and the ploidity of the NSCs and the co-cultured endothelial cells were scru- tinized, showed that six percent of adult NSCs transdifferentiated into endothe- lial like cells (Wurmser et al., 2004). In conclusion, only a small fraction of stem cells fuse with other cells; how- ever, this ability calls for extra caution and highlights the importance for con- trolled experimental set-ups. Before discussing NSC proliferation and fate determination, it is important to understand how the nervous system develops.

11 Development of the Nervous System Formation of Brain and Spinal Cord Neurulation is the stage in early neural development where dorsal ectoderm forms the neural plate, which in turn is closed to form the neural tube (Fig3). The neural ectoderm develops into the nervous system, and is also the source for neural stem cells. This stage of neural development is characterized by rapid proliferation, thus commonly referred to as the expansion phase (Panchision and McKay, 2002). This expansion in cell number is important and makes the foun- dation for the nervous system. For proper development of the brain, precursors must establish their posi- tional identity along the dorsal-ventral, anterior-posterior and left-right axis of the neural tube (Fig3). This positional identity is largely the result of morpho- gens secreted from adjacent tissues. For establishment of the anterior-posterior axis, factors such as basic fibroblast growth factor (bFGF) (Lamb et al., 1995), Wnt (McGrew et al., 1995) and retinoic acid (RA) (Papalopulu et al., 1996) are important, whereas for the dorsal-ventral axis, bone morphogenetic proteins (BMPs) and sonic hedge hog (Shh) are instrumental (Fig3). This patterning results in the anterior part of the neural tube forming the brain, and the posterior the spinal cord.

Neural ectoderm Dorsal Neural crest cells Posterior BMPs bFGF (nc) RA nc Wnt np

Neural fold (nc) Right Left nt

np Anterior Shh RA Ventral Figure 3. Neurulation and establishment of positional identity of the neural tube. nt, neural tube; np, neural plate; nc, neural crest. Once the stem cells of the neural tube have established their positional iden- tity, they will start to migrate and form different parts of the nervous system. The cortex is developed from the inside out, where young neurons migrate from the ventricular zone to the cell layers of the cortex where they form different types of mature neurons (Rakic et al., 1974; Nadarajah and Parnavelas, 2002). These migrating neurons are guided by radial glial cells, an elongated cell type that is in contact with both the ventricular zone and the pial surface of the devel- oping cortex (Fig4). Radial glial cell were previously thought to be of deter-

12 mined astrocytic lineage, but recent data propose that radial glia can give rise to new neurons and glial cells, suggesting that radial glia might have precursor properties (Parnavelas and Nadarajah, 2001; Rakic, 2003).

Pial surface Cerebellum Cerebral cortex Young neurons

Ventricle Ventricle Radial Spinal cord glia

Figure 4. Radial glial cells and migrating young neurons during formation of the cortex.

Neural Stem Cells In the developing embryonic brain, neural stem cells can be isolated from vari- ous regions such as the striatum, hippocampus and cortex. These NSCs can be cultured either as neurospheres (Fig5A), or as monolayer cultures (Fig5B). In either case, neural stem cells require either bFGF, or epidermal growth factor (EGF) to proliferate and to maintain their potency.

C DG

SVZ OB RMS

Figure 5. Cultured NSCs and neurogenic regions of the adult rodent brain (A) neuro- sphere (B) mono layer culture (C) DG, dentate gyrus; SVZ, sub ventricular zone; RMS, rostral migratory stream; OB, olfactory bulb. In the adult brain, neurogenesis occurs only in discrete areas such as the subven- tricular zone (SVZ) or the DG of the hippocampus (Altman and Das, 1965; Altman, 1969; Eriksson et al., 1998; Gould et al., 2001); as a result, these are the two regions from which adult NSCs can be isolated. The identity of the progenitors found in the SVZ is at the present moment under debate. One theory proposes that the nestin expressing ependymal cells present in the lining of the lateral ventricle are NSCs (Johansson et al., 1999). Opposing evidence is suggesting that SVZ neural stem cells are multipotent astrocytes that can form neurons in vivo (Chiasson et al., 1999; Doetch et al.,

13 1999; Laywell et al., 2000). These SVZ progenitors migrate as committed neuroblasts through the rostral migratory stream (RMS) to the olfactory bulb (OB) where they differentiate into interneurons (Fig5C) (Garcia-Verdugo et al., 1998; Alvarez-Buylla and Lim, 2004). Since there are no radial glial cells pre- sent in the RMS, it is unknown how these neuroblasts know where to migrate (Garcia-Verdugo et al., 1998).

The life of a Neural Stem Cell Neural stem cells are the most immature cells in the nervous system. These cells are going through both symmetric and asymmetric cell division, thus are both self renewing and multipotent (Temple, 2001). While symmetric stem cell divi- sion results in an exponential increase in cell number, asymmetric cell division maintains the stem cell pool and gives rise to more restricted progeny (Fig6). To ensure normal development and function of the nervous system, the brain must be of correct size and contain the appropriate cell number. In a normal neural tube of embryonic day (E) eight, E8, mouse embryos more than 50% of the cells are NSCs, while at post natal day (P) one, P1, less than 1% in the SVZ are left (Kalyani et al, 1997; Kalyani et al, 1998). During these approximately twelve days, most NSCs have turned into more restricted progeny as defined by McKay: Precursors, progenitors and post mitotic cells (Fig6) (McKay, 1997).

NSC

NSC NSC

NSC NP Transdifferentiation

Np Hepatocyte Myotube Figure 6. Life of a Gp NSC. NP, Neural Precursor; Np, Neu- ral progenitor; Gp, Astrocyte Oligo- Neuron Glial progenitor. dendrocyte While regulation of neural stem cell proliferation and fate determination is far from being completely understood, it is clear that a complex interplay between cell cycle regulators, important developmental path-

14 ways such as the Notch-Numb pathway, and cell fate specific transcription fac- tors are involved. These regulators will be dealt with in more detail below.

The Cell Cycle Because of the importance of correctly duplicating and transmitting DNA to daughter cells, the cell cycle is a highly regulated process. The cell cycle is di- vided into different phases. The G1 phase is the time between M- and S-phase, when the cell is respon- sive to mitogens. The G2 phase is important judged from a DNA integrity stand- point, since the replicated DNA is checked for accuracy before transmission to daughter cells. To ensure genome integrity, so called checkpoints involving a complex set of molecular interactions are activated late in G1 and at the G2/M interface of the cell cycle (Fig7).

G0 p16 p27 cdk4/6 M Cyclin D cdk1 Checkpoint Cyclin A/B G1

p21

cdk2 cdk2 Cyclin A/B Cyclin E p27

Restriction G2 cdk2 Cyclin A point S Checkpoint

Figure 7. The cell cycle and its most important cdk regulators.

Cell Cycle Regulators Cyclin Dependent Kinases The core cell cycle machinery is made up of the cyclin dependent kinase (cdk) complex which contains a catalytic protein, the cdk, and a regulatory compo- nent, the cyclin. For the cdk complex to be active, the cyclin must bind the cdk. Also, the cdk must be phosphorylated at one site, while dephosphorylated at another (Fig8).

15 in CyclinCycl Cyclin Cyclin cdk kinases phosphatases Cyclin inactive cdk cdk inactive inactive cdk P P active P P P

Figure 8. Regulation of cdk activity. Regulation of cdk activity is accomplished by periodic synthesis and degrada- tion of the cyclins (Hunt, 1991; Nurse, 2002), phosphorylation by kinases from the cdk-activating kinase family and dephosphorylation by phosphatases belong- ing to the family of cell division control (cdc) proteins. When active, the cdk complex will phosphorylate other cell cycle regulatory proteins. In addition, there are inhibitors of the cdk complexes. These inhibitors are di- vided into classes depending on both structure and cdk specificity. The four members (p15, p16, p18 and p19) of the inhibitors of cdk4 family will inhibit the catalytic subunit of cdk4 and cdk6 (Hannon and Beach, 1994; Serrano et al., 1993; Guan et al., 1994; Hirai et al., 1995; Chan et al., 1995). The other class of cdk inhibitors belongs to the Cip/Kip family, which inhib- its the activity of cyclinD, E and A dependent kinases. The members p21, p27 and p57 (Gu et al., 1993; Harper et al., 1993; El-Deiry et al., 1993; Xiong et al., 1993; Dulic et al., 1994; Noda et al., 1994; Polyak et al., 1994; Toyoshima and Hunter, 1994; Matsuoka et al., 1995) will bind not only to the cdk subunit, but also to cyclins (Chen et al., 1995; Nakanishi et al., 1995; Warbrick et al., 1995; Lin et al., 1996; Russo et al., 1996).

Notch and Numb Pathway One of the best studied developmental pathways is the Notch and Numb path- way. The first clue that these proteins were important during early patterning came from localization studies, which revealed their asymmetric distribution in the cell (Chenn and McConnel, 1995; Zhong et al., 1996; Wakamatsu et al., 1999; Wakamatsu et al., 2000). The Notch protein is a transmembrane receptor that is interacting with both extra-cellular proteins on neighboring cells, and proteins within the cell. The extra-cellular activation of the Notch receptor is mediated by the transmembrane ligands Delta and Jagged (Mumm and Kopan, 2000; Weinmaster, 2000). Activa- tion of the Notch receptor results in a proteolytic cleavage of the intracellular Notch (ICN) domain, which acts as a transcriptional co-activator of the target genes Hairy and enhancer of split homolog (Hes) 1 and 5 (Fig 9A)(Furukawa et al., 2000; Lundkvist and Lendahl, 2001). Since Hes genes repress transcription of the important pro-neural genes Mash and Neurogenin (Ngn) 2, NSCs are kept in a proliferative state (Fig9A). The outcome of Notch signaling is repression of pro neural genes; thus, Notch signaling favors glial and radial glial fate (Gaiano et al., 2000).

16 However, the proteolytic cleavage of the ICN can be inhibited by Numb, a Notch antagonist, by binding to the secretase binding site (Fig9B) (Verdi et al., 1996; Wakamatsu et al., 1999; Zhong et al., 1996). There are four alternatively spliced forms of the human Numb (Verdi et al., 1999). Interestingly, two of these splice forms induce proliferation while the other two mediate neuronal fate determination (Verdi et al., 1999).

A B

DELTADEL DELTADEL NOTCHNO NOTCH T TA A TC H

CoR Hes Ngn, Mash activation PS NUMB NUMB ICN

ICN Activation PS CoR Repression TF TF Notch responsive genes (Hes1,5) Notch responsive genes (Hes1,5)

NP NP

Ngn Ngn

Ngn Ngn Neuron Neuron

Astrocyte Astrocyte Figure 9. Notch signaling in neural precursors (NP).

(A) Proteolytic cleavage of ICN. (B) Numb inhibition of ICN signaling. TF, ; PS, presenillin (secretase which can cleave of ICN); CoR, co-repressor.

Cell Fate Specific Transcription Factors: the bHLH Proteins Initially, there is a balance between Notch signaling and the expression of basic helix loop helix (bHLH) transcription factors that either inhibit or promote dif- ferentiation. Still, at some point, the stem cell must exit the cell cycle and be- come post mitotic. This regulation of mitotic exit is, to a large extent, the result of some early bHLH pro-neural proteins (Ohnuma and Harris, 2003; Kintner, 2002). These bHLH transcription factors will, in a cascade like manner, activate other downstream bHLH neuronal determination genes in the neuro-epithelium (Ma et al., 1996). When the protein levels of these neuronal determination genes

17 has reached a thresh hold, cdk inhibitors are activated (Ohnuma and Harris, 2003), and the stem cell will exit the cell cycle. Of course, the regulation of mitotic exit is much more complicated. Cross talk and intricate feedback loops connect the bHLH transcription factors with Notch signaling and the cell cycle. But what system has the decisive power? Recently, studies have shown that the determinant power is in the hands of the bHLH proteins (Ohnuma and Harris, 2003). For example, both activation of the Notch receptor and an increase in the cdk inhibitor p27, will independently force the cell to become post mitotic (Scheer et al., 2001; Ohnuma et al., 1999). On the other hand, when pro-neural bHLH proteins are co-expressed with Notch or p27, the bHLH proteins will decide the outcome (Ohnuma et al., 2002). In addition to these regulatory systems, mitogens (which are discussed in the context of fate determination) and cell adhesion molecules such as integrins are also important regulators of proliferation. Integrins mediate signals from the exterior to the interior of the cell by binding to extra-cellular matrix proteins such as laminins or other integrins located on other cells. As a result, integrin signaling activates intracellular signal pathways such as phosphatidylinositol-3 (PI-3) and mitogen activated protein (MAP) kinase pathways.

Differentiation and Fate Determination

The G1 Restriction Point For a neural stem cell to start to differentiate and become post mitotic, it must enter G0 without passing the G1 restriction point (Fig7, 10). This restriction point is primarily regulated by cdk4, 6 and 2, cyclinDs, Es, Retinoblastoma protein (pRb), p53, and the cdk inhibitors (Fig10). If cells are blocked in G1, by for example over expression of p27 or down regu- lation of cyclinE1/D1, the bHLH determination pathway is activated. The cells then enters G0 and becomes post mitotic (Ohnuma et al., 2002; Ratineau et al., 2002). Interestingly, in the hippocami of P5 mice, the mRNA for all three cyclins (cyclinD1, D2 and D3) are present, while in proliferating precursors of the adult hippocampus only cyclin D2 is expressed (Kowalczyk et al., 2004). This illus- trates that the cell cycle in developmental and adult precursors are differentially regulated.

18 p130 Repression G0 E2F responsive genes (c-, cdc2, )

pRb Repression E2F G1 E2F responsive genes (c-myc, cdc2, E2F1)

cdk4/6 p15 Cyclin D p16 P P p53 P cdk2 p21 pRb Cyclin E Figure 10 . Regula- P P p27 tion of the G1 restric- P tion point. p130 cdc25 (member of the pRb family); cdc, cell P Activation division control E2F protein. S E2F responsive genes (c-myc, cdc2, E2F1)

Cell Fate Specific Transcription Factors: bHLH Proteins During neural development, neurogenesis occurs before gliogenesis. However, both neurons and glial cells are derived from the same neuro-epithelium. Inter- estingly, the same core program of bHLH transcription factors regulates NSC proliferation as well as neurogenesis and gliogenesis (Kintner, 2002). This large family of transcription factors contains both pro-neural bHLH pro- teins such as Mash1, Ngn1 and 2, NeuroD and Math3, as well as transcription factors which direct progenitors into a gliogenic fate such as Hes1 and 5, and 2, and Ngn3 (Kintner, 2002). Initially, bHLH proteins such as Hes, Mash1 and inhibitor of differentiation (Id), are turned on by the early patterning morphogens BMPs and Shh (Sau- vageot and Stiles, 2002; Morrison, 2001; Panchision and McKay, 2002). These bHLH transcription factors in turn induce expression of other bHLH transcrip- tion factors, initiating a bHLH cascade (Ma et al., 1996; Kintner, 2002). When the protein levels of key bHLH transcription factors pass a thresh hold level, cdk inhibitors are turned on, and the cell is forced out of the cell cycle in favor of a differentiated phenotype. Some of the known bHLH transcription factors that are instrumental in NSC fate determination are summarized in Fig11.

19 NSC bFGF, EGF

bHLH proteins Mitogens

Mash1, Ngn1,2 BMP2,4 NeuroD, Math3 PDGF, bFGF Figure 11. bHLH and mitogens in- Neuron Neuron volved in NSC fate determination. Hes1,5, Id LIF, CNTF, Ngn3, Sox9 BMP2, Notch* * Notch is neither a bHLH transcrip- Astrocyte Astrocyte tion factor nor a Olig1,2, mitogen, but in- Id2, Nkx 2.2 strumental in NSC Sox 8,10 Shh, T3 fate determination.

Oligodendrocyte Oligodendrocyte

Mitogens As previously discussed, mitogens are important during early neural develop- ment when BMP and Shh aid the patterning of the neural tube. While the deter- minant power as far as regulating NSC fate determination is in the hands of intrinsic regulators, a handful of important extrinsic factors have also been iden- tified. It has been estimated that proliferating NSCs in vivo will undergo 10-12 cell divisions before becoming post mitotic (Takahashi et al, 1994). In vitro, NSCs isolated from the early brain are dependent on bFGF for proliferation (Kalyani et al., 1997; Raballo et al., 2000; Vaccarino et al., 1999), while at later stages NSCs require either bFGF or EGF for proliferation (Gritti et al., 1999; Tropepe et al., 1999). Upon withdrawal of EGF or bFGF, the NSCs will spontaneously differentiate into neurons and glial cells (Johe et al., 1996). A number of mitogens have been identified that can influence the fate determination upon EGF or bFGF with- drawal, and some of the most potent ones are summarized in Fig11 and will be discussed below. Platelet derived growth factor (PDGF) is primarily known to support neu- ronal differentiation of both embryonic and adult NSCs (Johe et al., 1996; Wil- liams et al., 1997), probably not by instructing the NSCs, but rather by expand- ing the pool of neural precursors (Erlandsson et al., 2001). Another potent mitogen that regulates fate determination is the ciliary neuro- trophic factor (CNTF), which will induce astroglial fate (Johe et al., 1996; Bon- nie et al., 1997). The thyroid hormone tri-iodo-thyronine (T3) will induce glial

20 fate and increase the formation of oligodendrocytes and astrocytes (Johe et al., 1996). The bone morphogenetic proteins involved in neural tube patterning increase the formation of neurons in E12 NSCs (Li et al., 1998). However, the outcome of bone morphogenetic protein (BMP) regulated fate determination depends on the age of the stem cell. For instance, cortical E13 precursors will undergo apop- tosis if stimulated with BMP4, while E16 NSCs primarily differentiate into neu- rons and glial cells, and postnatal NSCs primarily form glial cells (Gross et al., 1996; Mehler et al., 2000). Another example of differential fate regulation is the treatment of E12.5 cor- tical precursors with CNTF or leukemia inhibitory factor (LIF), which does not affect fate determination (Molne et al., 2000), while the same treatment will instruct E14.5 cortical precursors to form 98 % astrocytes (Johe et al., 1996). Considering the importance of the bHLH proteins in regulation of NSC fate, it is not surprising that the most potent mitogens affect these transcription fac- tors (Fig12). PDGF

Shh BMP CNTF BMP CNTF Delta

ICN Olig Smad Ngn CBP/p300 CBP/p300 Hes Id Ngn STAT STAT ASTRO NEURO NEURO ASTRO DETERMINATION DETERMINATION DETERMINATION OLIGO DETERMINATION GENES GENES GENES DETERMINATION GENES GENES NEURO ASTRO NEURO OLIGO ASTRO PROMOTOR PROMOTOR PROMOTOR PROMOTOR PROMOTOR

Figure 12. Crosstalk between mitogens and downstream transcriptional regulators. Astro, astrocytic; Neuro, neuronal; Oligo, oligodendrocytic. CBP/p300, general co- activator for multiple signaling pathways.

Mature Cell Types within the Central Nervous System

Neurons Neurons are unique in the sense that they are highly specialized for inter-cellular signaling. As a result, neurons have a special morphology, membrane formation and the ability to form synapses with connecting neurons.

21 The axon is optimized for signal transduction, and its length can vary be- tween neuronal types depending on how far the signal needs to be transmitted. While axons transmit information, dendrites are specialized for receiving infor- mation. As in the case of axons, the extension and branching of the dendrites vary between neuronal types.

Figure 13. Neuron, astrocyte and oli- godendrocyte formed from embryonic NSCs. Size bar 20Pm.

Glial cells The most abundant cell types in the nervous system are the glial cells. There are three types of glial cells in the central nervous system (CNS): astrocytes, oli- godendrocytes and microglia. Glial cells have important supportive roles which include for example provid- ing neurons with trophic support; as a result, their functions have previously been underestimated. Recently, a study showed that factors secreted from astro- cytes are able to instruct NSCs into forming neurons (Song et al., 2002). Also, Alvarez-Buylla and co-workers have identified a population of proliferating glial fibrillary acidic protein (GFAP) positive precursors in the vicinity of the lateral ventricle of the adult brain (Seri et al., 2001). At the present moment, evidence is pointing towards that multipotent astrocytes are in fact the SVZ NSC which can form neurons in vivo. However, astrocytes also have negative roles on the nervous system, as in the case of nerve regeneration. After axonal damage, astrocytes form glial scares which constitute a mechanical and chemical barrier for axon sprouting (Emsley et al., 2004).

22 In order to enhance the speed at which information is transferred between neurons, their axons are myelinated by the oligodendrocytes which are found only in the CNS. The microglial cells are the only CNS cell type that is not derived from NSCs but rather from the hematopoetic system. Upon damage, microglia will prolifer- ate, and perform macrophage like functions.

Markers Used in NSC Research Because studies of the nervous system depend on the ability to accurately distin- guish between different cell types, biological markers are used. Some of these markers are structural proteins, such as nestin, E-III-tubulin and GFAP, while others are transcription factors such as Sox1. Some of the most commonly used markers in NSC research are summarized in Table1.

Table 1. Cell markers commonly used in NSC research. Cell type Marker Reference

NSCs Nestin Lendahl, 1990 ” Musashi Yagita et al., 2002; Sakakibara et al., 2002 ” Sox1, 2 Pevny et al., 1998; Sasai, 2001 ” Bmi-1 Molofsky et al., 2003 ” Vimentin Houle and Fedoroff, 1983 Immature Neurons E-III-tubulin Caccamo et al., 1989 ” MAP-2 Garner et al., 1988; Matus et al., 1988 Immature glial cells A2B5 Hirano and Goldman, 1988 ” Olig 1 Zhou et al., 2000. ” Sox 10 Kuhlbrodt et al.,1998 ” Nkx 2.2 Qi et al., 2001 ” Ngn 3 Liu et al., 2002 Neurons NeuN Mullen et al., 1992 ” NF (Neurofilament) Debus et al., 1983 Cholinergic ChAT Haigh et al., 1994 Dopaminergic TH Nagatsu et al., 1964 GABAergic DARPP-32 Lewis et al., 1983 Glutamatergic VGLUT1, 2 Arriza et al., 1994 Seratonergic SERT Blakely et al., 1991 Astrocytes GFAP Eng et al., 2000 ” S100-E Zimmer at al., 1995 Oligodendrocytes CNPase Staugatis et al., 1990 ” O4 Sommer and Schachner, 1981 ” MBP Hartman et al., 1982

23 Hormones

Sex Hormones Sex hormones are structurally related substances, which are formed from the common precursor cholesterol (Fig14). These hormones are crucial in sexual differentiation and development of the reproductive system. The most potent member of the estrogens, 17E-estradiol, what will be referred to as estrogen, is primarily synthesized in the ovaries of pre-menopausal women, but also locally in fat tissue and the brain. Females that carry a loss of function mutation in the cytochrome P450-19 enzyme, aromatase, responsible for con- verting testosterone to 17E-estradiol, have disturbed sexual differentiation and as a result male characteristics. The male sex hormones are both androgenic, that is, responsible for devel- opment of male characteristics, and anabolic, promoting protein synthesis. Sub- stances which promote both the anabolic and the androgenic function are re- ferred to as anabolic androgenic steroids (AAS). Among the AAS both synthetic testosterone analogues such as 19-nortestosterone commonly referred to as nan- drolone, and the in testis formed endogenous hormone testosterone are found. Testosterone, and its more potent metabolite dihydotestosterone (DHT), as well as nandrolone bind with high affinity to the androgen receptor (AR), and acti- vate transcription of AR responsive genes (Deslypere et al., 1992; Roselli, 1998). Males with mutated androgen receptor are insensitive to androgens, and thus develop female characteristics.

24 Cholesterol

HO

Pregnenolone Progesterone

Corticosteroids 17α-OH-Pregnenolone Dehydroepi- Androsterone

H CF3 α 17 -OH-Progesterone N NO2 O Flutamide Androstenedione

N

OH OH O OH Tamoxifen

Aromatase OH O O HO Testosterone 19-Nor- 17E-Estradiol O testosterone S (Nandrolone) CF3 Dihydro- HO Testosterone- ICI 182780 (DHT)

Figure 14. Steroid biosynthesis and some NR antagonists. Flutamide (AR antagonist), Tamoxifen (ERD, ERE mixed antagonist/agonist), ICI 182 780 (pure ERD and ERE antagonist).

17E-Estradiol Estrogen Receptors 17E-Estradiol acts by binding to estrogen receptors (ERs) of which two types, ERD and ERE are known (Green et al., 1986; Greene et al., 1986; Kuiper et al., 1996). These hormone receptors belong to the super family of nuclear receptors (NRs). NRs have four functional domains including a ligand independent N- terminal activation function (AF) domain, a central DNA binding domain con- sisting of two zinc fingers, a hinge domain and a C-terminal ligand-binding domain. Within the ligand binding domain there are also motifs for receptor dimerization, nuclear localization, and transactivation (Fig15).

ERα NH3 DNA Ligand COOH

AF-1 hinge dimerization AF-2

ERβ NH3 DNA Ligand COOH

Figure 15. Structural motifs of ERs (example of classical NR).

25 The physiological response of estrogen in cells is primarily mediated by binding of the hormone/receptor complex to target DNA, which leads to effects on gene transcription. This genomic response requires transcription and transla- tion, and cannot explain the extremely rapid estrogen exerted modulation of plasma membrane bound neurotransmitter receptors (Gu et al., 1999), calcium currents (Mermelstein et al., 1996; Beyer and Raab, 1998; Carrer et al., 2003) and G protein coupled receptors (Qiu et al., 2003). As a result, there are two separate actions mediated by ERs. First, the classi- cal genomic action, which are on the time scale of hours and can be abolished by transcriptional inhibitors. Second, the non-genomic action, which is rapid (sec- onds-minutes), and is completely insensitive to transcriptional inhibitors (Beyer et al., 2003). In the brain, ERE is the primary ER; however, both receptors have largely overlapping distribution with highest densities in the forebrain, hypothalamus, amygdala and septum (McEwen, 2001). Different splice variants have been found for both ERD and ERE and they are differentially expressed in various regions with possible functional consequences (Shughrue et al., 1997; Gundlah et al., 2000; Carroll et al., 1999; Patrone et al., 2000). In particular, in regions of the substantia nigra and cerebellum, ERE the exclusive isoform, whereas ERD is seen in the ventromedial hypothalamic nucleus (Shughrue et al., 1997). Interest- ingly, mRNA for estrogen receptors are found in cultured glial cells (Santagati et al., 1994), but in the adult brain, ERE seem to be the only ER expressed by astrocytes (Azcoita et al, 1999). In addition to the differential expression, ERs are able to further diversify their response following hormone binding by form- ing homo- and hetero-dimers.

Effects of Estrogen in the Brain Besides the effects on the reproductive system, estrogen exhibits neurotrophic properties promoting growth, survival and maintenance of neurons during de- velopment of the nervous system leading to sexual differentiation of the brain (McEwen, 1983; MacLusky et al., 1987; Toran-Allerand, 1991). Also, estrogen influences synaptogenesis and contributes to synaptic plasticity (McEwen et al., 2001). In particular, estrogen enhances growth and differentiation of neurons during development (Toran-Allerand et al., 1999), stimulates neurite outgrowth of hy- pothalamic neurons (Ferreira et al., 1991) and increases dendritic length of em- bryonic neurons from the medial amygdala (Lorenzo et al., 1992). In neurons from the adult female rat hippocampus, estrogen alters the morphology as well as increases the number of synapses and density of the dendritic spines (Woolley et al., 1990; Gould et al., 1990; Woolley and McEwen, 1992). In addition, estrogen also exhibits neuroprotective effects by modulating pro- tein levels of anti- and pro-apoptotic proteins. For instance, estrogen protects hippocampal and dopaminergic neurons from apoptosis by up regulating Bcl-xl and Bcl-2 expression (Pike, 1999; Sawada et al., 2000). Interestingly, estrogen

26 increased the ratio of ERE/ERD after ischemia (Paech et al., 1997). The ERD knockout (ERKO) mouse did not show increased sensitivity against ischemic insult compared with wild type littermates (Sampei et al., 2000). This has led to the idea that the most important ER isoform for neuroprotection is ERE. Also, estrogen has shown positive effects on the neurotransmitter systems in- volved in Alzheimer’s and Parkinson’s disease such that estrogen increases the choline acetyl transferase activity in the female rat hippocampus (Luine, 1985) and regulates the survival of dopaminergic neurons in the substantia nigra of the monkey brain (Leranth et al. 2000). As a result, the density of dopaminergic neurons is higher in female brain than male (Leranth et al. 2000). While pre-menopausal women have a lower incidence of neurodegenerative disorders than men or post menopausal women, results from clinical trials where hormone replacement therapy (HRT) was given are conflicting. For instance, Beral report a substantial incidence in breast cancer (Beral, 2003), while Shumaker and colleagues report an increase in probable dementia after HRT (Shumaker et al., 2003). One interesting explanation for the inter-individual variance in following HRT in humans is the genetic variance in apolipoprotein E (apoE) expression (Lehtimaki et al., 2002; MacLusky, 2004). ApoE has, like- wise estrogen, showed neurotrophic and neuroprotective effects (Nathan et al., 2002). Since the ability of estrogen to mediate neuroprotection is abolished in transgenic mice bearing an apoE mutation, the trophic and protective effects of estrogen might be mediated by apoE (Horsburgh et al., 2002).

ER Dependent Cell Signaling Estrogen is known to cross-couple many important signaling pathways. For instance in human retinoblastoma cells, insulin activated the ER via involvement of the AF-2 domain (Patrone et al., 1996). Also, estrogen turns on the cAMP/CREB (Hanstein et al., 1996) and the PI-3 kinase/Akt pathway (Simon- cini et al., 2000). By directly regulating c-myc and cyclinD1 levels, estrogen exerts profound effect on the cell cycle (Doisneau-Sixou et al., 2003). Some of the signaling pathways which are activated by estrogen affects are outlined in Fig16.

27 NON GENOMIC RESPONSE Ion channels Gs/Gq coupled receptors E2 E2 E2 ER ER E2 E2 E2 ER ER E2 Neurite outgrowth

E2 synaptic plasticity hsp90 Ca2+ E2 ER hsp90 MAPkinase PI3 kinase E2 cAMP-PKA

Cytoskeleton hsp90 Transcription E2ER ERE2 E2 ER ER E2 E2

E2 E2 hsp90 hsp90 E2 ER GENOMIC hsp90 RESPONSE anti apoptotic proteins

E2 pro apoptotic proteins E2 Growth factor Neurotransmittor receptors modulation Protection, survival

Figure 16. Genomic and non genomic responses of estrogen signaling

19-Nortestosterone Abuse of anabolic steroids have become more common among adolescents and extended outside of the sphere of elite athletes and body builders (Kindlundh et al., 2001). Abuse of nandrolone is known to result in behavioral changes mainly associated with increased irritability and aggression (Lynch and Story, 2000; Johansson-Steensland et al., 2002). Also, problem with the maintenance of spermatogenesis and increased masculinization/feminization is reported (Lynch and Story, 2000; Johansson-Steensland et al., 2002).

Actions in the Brain Little is known about androgenic anabolic steroids in the brain. However, re- cently AAS has been shown to decrease serotonin levels and alter serotonin receptor levels (Lindqvist et al., 2002; Kindlundh et al., 2003), dopamine recep- tor density (Kindlundh et al., 2001) and expression levels of N-methyl-D- aspartate receptor subunits (Le Greves et al., 1997). AAS treatment also up- regulates the androgen receptor (Menard and Harlan, 1993), and increases the density of fos-like immuno reactive cells (Johansson-Steensland et al., 2002), suggesting that AAS can stimulate different brain regions.

Androgen Receptor 19-Nortestosterone acts by binding to its receptor, the androgen receptor, which belongs to the super family of nuclear receptors (Heinlein and Chang, 2002a). The androgen receptor preferably forms homodimers, but is also known to het- erodimerize with other NR such as ERD and the . Just as in

28 the case of ERs, AR is exerting both fast non genomic and genomic responses, which require transcription and translation (Heinlein and Chang, 2002b). In the brain, the androgen receptor has been localized to neurons (Finley and Kritzer, 1999) and glial cells (Hösli et al., 2001) in areas mainly associated with reproductive behavior such as the hypothalamus. However, the AR is also found in the lateral septum, stria terminalis, pre-optic area and in the medial amygdala (Lynch and Story, 2000).

Sex Hormone Knock Outs In order to study the effects of sex hormones during development, in different organs and during aging, mouse models have been prepared. At the present mo- ment there are knockouts for both ERD (ERKO), ERE (BERKO), ERD/E (ERKO +BERKO), AR (ARKO) and the enzyme aromatase converting testos- terone to estrogen (ArKO). These knock outs all have some defects in reproduc- tion, while the BERKO mouse in particular have effects on the nervous system. The major phenotypes are summarized in Table 2.

Table 2. Sex hormone related mouse models. Model Fertility Phenotype Reference

ERKO Ƃ infertile Defected uterus, mammary gland and Lubahn et al., 1993; ƃ infertile pituitary. Bocchinfuso et al., 1997; No brain defects. Couse et al., 1999; Couse and Korach, 1999.

BERKO Ƃ Ļ fertility Ovarian defects. Krege et al., 1998; Wang ƃ normal fertility Smaller brains, fewer neurons, et al., 2001; Wang et al., Ĺastroglial proliferation, 2003. disturbed neuronal migration in cor- tex,Ĺapoptosis

ERKO/ Ƃ infertile No ƃsexual behavior. Ogawa et al., 2000. BERKO ƃ infertile

ArKO Ƃ infertile Disturbed sexual behavior. Fisher et al., 1998; Ma- ƃ Ļfertility Decreased aggressiveness in male. tsumoto et al., 2003

ARKO Ƃ infertile ƃfeminization, smaller testicles, Yeh et al., 2002. ƃ Ļ fertility adipocyte alterations.

The Melanocortin System The melanocortin family consists of the peptides adrenocorticotrophic hormone, D-, E-, and J- form of the melanocortin stimulating hormone, and the two en- dogenous melanocortin receptor (MCR) antagonists agouti and agouti related

29 protein. The melanocortins are formed by post translational processing of the precursor peptide pro-opio-melanocortin (POMC) by the pro-convertase 1 and 2 (Nakanishi et al., 1979; Smith and Funder, 1988; Benjannet et al., 1991). This processing is tissue specific (Pritchard et al., 2002). Melanocortins have roles in the regulation of many aspects of human physi- ology such as thermoregulation (Feng et al., 1987), obesity (Fan et al., 1997), skin pigmentation (Thody, 1999), and anti inflammatory responses (Catania et al., 2000; Luger et al., 1999).

Melanocortins in the Brain In the brain, melanocortins have stimulatory effects on learning and memory (De Wied and Croiset, 1991), but also on neural outgrowth in for instance stri- atal and mecencephalic cells via D-MSH activation of MC4R (Kistler-Heer et al., 1998), in foetal spinal cord neurons (Van der Neut et al., 1988), in postnatal sensory neurons (van der Neut et al., 1992) and in retinal neurons (Lindqvist et al., 2003). The precursor POMC is expressed in the CNS in particular in the pituitary, hypothalamus and areas of the brain stem (Gantz and Fong, 2003). Five Gs-protein coupled melanocortin receptors (MC1R-MC5R) have been cloned which are differentially expressed and have varying affinity for the melanocortins (Gantz and Fong, 2003). The peptide D-MSH has the highest affinity for MC3R and MC4R (Wikberg, 1999), which are expressed in the CNS especially in the areas of hypothalamus and thalamus where they are involved in energy homeostasis (Gantz et al., 1993; Lindblom et al., 1998; Xia and Wikberg, 1997). MC3R is in addition abundantly expressed in the septum, hippocampus and midbrain (Roselli-Rehfuss et al., 1993; Xia and Wikberg, 1997). MC4R is ubiquitously distributed in almost all regions of the mammalian brain, including spinal cord, brain stem and cortex (Gantz et al., 1993). Melanocortin receptors are primarily localized to neurons, but can also be found in astrocytes as well (Wong et al., 1997). D-MSH dependent activation of MC4R in the hypothalamus affects energy homeostasis, which leads to a diminished feeling of hunger and a subsequent decrease in food intake (Fan et al., 1997).

BRCA-1 The breast cancer susceptibility gene one, BRCA-1, was first identified as a candidate gene involved in heritable breast and ovarian cancer (Miki et al., 1994). Germline mutations in the two breast cancer susceptibility genes, BRCA- 1 and BRCA-2, are responsible for two thirds of familial cases of breast cancer (Alberg et al., 1998).

30 Using cultured breast and ovarian cancer cell lines as well as hypomorphic and tissue specific mutant mice, BRCA-1 has been shown to be involved in DNA damage repair, centrosome duplication, cell cycle arrest, growth retarda- tion, apoptosis, genetic instability and tumerogenesis (Brodie and Deng, 2001; Welcsh et al., 2000).

BRCA-1 Structure The human BRCA-1 is an 1863 amino acid large protein with structural motifs that reveal its important functions (Fig17). The N-terminal part of the protein contains a ring domain. Many important cell cycle regulators have been found to interact directly or indirectly with the N-terminal part of BRCA-1 such as: p53 (Chai et al., 1999; Zhang et al., 1998; Ouchi et al., 1998;), pRb (Aprelikova et al., 1999), RAD51 (Scully et al., 1997a; Zhong et al., 1999; Scully et al., 1997c), E2F1 (Wang et al., 1997), BARD1 (Wu et al., 1996) and c-myc (Wang et al., 1998). Located in the central part of BRCA-1 are two nuclear localization sig- nals which bind to the transport receptor importin-D, enabling transport over the nuclear envelope. The C-terminal part of BRCA-1 contains two BRCA-1 C- terminal repeats (BRCT) that interact with proteins such as p53 (Ouchi et al., 1998; Chai et al., 1999), RNA pol II (Scully et al., 1997b; Neish et al., 1998), pRb (Yarden et al., 1999) and BRCA-2 (Chen et al., 1998). These BRCT do- mains are found in many DNA repair and cell cycle check point proteins (Bork et al., 1997; Callebaut and Mornon, 1997; Koonin et al., 1996).

Transcriptional NLS activation DNA binding BRCT Figure 17. Struc- NH3 RING COOH tural motifs of BRCA-1 and some E2F1 p53 α-importin of its cell cycle c-myc p53 RAD50 BARD1 Rb BRCA-2 RAD51 RNApol II interactors.

BRCA-1 Expression During development, BRCA-1 mRNA expression is detected as early as in the E6.5 mouse, with a peak at E13.5 (Rajan et al., 1997). The highest levels of the BRCA-1 protein are observed in tissues containing rapidly proliferating cells, in particular in those undergoing differentiation such as the mammary epithelium (Rajan et al., 1996; Lane et al., 1995; Marquis et al., 1995). During development of the nervous system, BRCA-1 is expressed in the wall of the lateral ventricle, but also in the forth ventricle and areas of the midbrain (Rajan et al., 1997). In the adult, BRCA-1 is primarily expressed in sex hormone responsive tissues such as testis and thymus (Miki et al., 1994; Rajan et al., 1997).

31 During the cell cycle, both BRCA-1 mRNA and protein levels are low during G0 and G1, but obtain maximum levels at the G1/S phase transition (Chen et al., 1996; Ruffner and Verma, 1997). BRCA-1 becomes hyperphosphorylated dur- ing the G1/S interphase and forms nuclear aggregates which can be seen as dis- tinct nuclear dots (Ruffner and Verma, 1997; Scully et al., 1997a; Scully et al., 1997c). This phosphorylation is especially apparent in cells which have been subjected to DNA damage, when BRCA-1 co-localizes with other DNA damage proteins such as BARD1 and RAD51, forming a multiprotein complex which plays a part in the replication checkpoint response (Scully et al., 1997c).

BRCA-1 Mutant Mice In order to study the effect of BRCA-1 in tumerogenesis in vivo, a number of different mouse models have been created. The BRCA-1 null mice are embry- onic lethal due to the proteins importance during early embryogenesis. However, mice homozygous for several of the familial mutations, and mouse models in which parts of the BRCA-1 gene is deleted. They die during early embryonic development (E8.5-E13) due to developmental delay and defective proliferation (Hakem et al., 1996; Liu et al., 1996; Ludwig et al., 1997; Hakem et al., 1998).

Estrogen Dependent Regulation of BRCA-1 Mutations in the BRCA genes are found in most familial cases of breast and ovarian cancer (Kerr and Ashworth, 2001), which are estrogen responsive tis- sues. One of the first potent treatments for breast cancer was tamoxifen (Fig14), a substance that binds to the ERs. Recently, molecular evidence for the tamoxifen action in BRCA-1 mutated cells was discovered by Zheng and colleagues who showed that BRCA-1 re- presses the transcriptional activity of ERD (Zheng et al., 2001). Also, estrogen stimulation in breast cancer cells results in a sustained activation of extra- cellular signal-related kinase (erk), which is abolished if wild type BRCA-1 is introduced (Razandi et al., 2004). Further, BRCA-1 interacts with the AF 2 do- main of ERD, regulating transcription of vascular endothelial growth factor (Fan et al., 2001; Kawai et al., 2002).

32 MATERIALS AND METHODS

Experimental Animals Time pregnant Wistar rats were purchased from B&K (Sollentuna, Sweden), and housed at 12h: 12h light: dark cycle with free access to food and water. All ex- periments were approved by the local ethical committee, and carried out in ac- cordance with the European Communities Council Directive (86/609/EEC).

Nandrolone/BrdU Injections Male, female, and time pregnant E15 Wistar rats (~260g) were subject to daily subcutaneous injections for five days of either nandrolone (15mg/kg body weight, Deca-Durabol, Organon, Netherlands) or vehicle (peanut oil, Apoteket AB, Umeå, Sweden). During the first three days, the rats received daily intrap- eritoneal injections of 5´-bromo-2-deoxy-uridine (BrdU) (100mg/kg body weight; Sigma). After five days of injections, rats were anaesthetized using chlo- ral hydrate (Apoteket AB, Umeå, Sweden) and perfused with 4% paraformalde- hyde (PFA) in phosphate buffered saline (PBS) pH 7.4, followed by post fixa- tion of the brains for 24 hours in PFA. The brains were dehydrated, embedded in paraffin and 10Pm sections were produced using a sliding microtome.

Cell Culture

Primary Neural Stem Cells Striatal tissue was dissected from E10-E20 Wistar rats, dissociated and prepared as described in paper I-IV. NSCs were cultured in medium consisting of 15 mM HEPES (pH 7.5), 2 mM L-glutamine, 100 U/ml penicillin, 100 mg/ml strepto- mycin and B27 supplement (Invitrogen) in DMEM-F12. At the beginning of incubation, 20ng/ml of EGF (Invitrogen) was added. Cells were incubated at 37qC in 5% CO2 atmosphere in non adherent Falcon dishes or Nunc flasks.

33 To prepare adult NSCs, the lining of the lateral ventricles of adult female Wistar rats were carefully dissected, dissociated and adult SVZ NSC were prepared as described before (Johansson et al., 1999), and cultured in the presence of both EGF (20ng/ml) and bFGF (20ng/ml).

Cell Biology

Western blotting NSCs were lysed in 62.5 mM Tris-HCl (pH 6.8), 2% SDS, 10 % Glycerol, and 50mM dithiothreitol in the case of the NR blots. In the case of other blots, a buffer containing 50mM Tris (pH 8.0), 150 mM NaCl, 1% TritonX-100 (TX- 100), 0.5% Natriumdeoxycholate, 0.1% SDS, 1 mM Sodium orthovanadate and Proteinase inhibitor (Roche) was used. Protein concentration was determined by BioRad protein assay DC (BioRad, Sweden) and equal amounts of proteins were loaded onto SDS-PAGE gel for separation. Proteins were transferred onto a polyvinylidene difluoride (PVDF) or nitrocellulose membrane and blocked in 3- 5% skimmed milk in TBS-T (50mM Tris pH 7.6, 150mM NaCl, 0.1% Tween- 20). Membranes were typically incubated overnight at +4qC with primary anti- bodies (Table3), followed by washing in TBS-T. Secondary horse radish peroxi- dase (HRP) conjugated antibodies (Pierce) were added for 2-4 hours in 2-5% skimmed milk in TBS-T at room temperature. The membrane was washed in TBS-T and bands were visualized using the ECL method (Amersham, Sweden).

Cell Proliferation

BrdU Assay To study proliferation, mechanically dissociated NSCs were placed in small 35 mm Petri dishes (500,000 cells/dish, Falcon, Sweden) in culture medium in the presence of factors of interest. After two days, a 12-24 hour pulse of 10PM BrdU was given. The neurospheres were dissociated and placed on Poly-DL- ornithine (Sigma) coated 48 well plates (BD Biosciences). The number of BrdU positive cells was analyzed by immunocytochemistry.

Flow Cytometry Flow cytometry was also used to count BrdU positive cells. Shortly, NSCs were pulsed with BrdU for 12 hours and fixed with 70% ethanol. DNA was denatu- rated using 2M HCl, rinsed in phosphate-citric acid buffer pH 7.4. Primary anti BrdU (1:100; M0744, Dako, Glostrup, Denmark) was added, followed by incu- bation with fluorescein isothiocyanate (FITC) labeled secondary antibody (1:200; F0313, Dako). The flow cytometric analysis was done using the fluores-

34 cence-activated cell sorting equipment, Calibur (BD Immunocytometry Sys- tems, San Jose, CA USA) with an analysis rate of 700-2000 cells/s collecting forward scatter, side scatter and green signals. Data analysis was done using the Cellquest software.

Cell Number/Viability Assay Total NSC number after different treatments were also studied using the 3-(4,5- dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay, which is based upon the reduction of MTT by mitochondrial hydrogenases. Briefly, NSCs (500,000 cells/dish; Falcon), were incubated for three days under various conditions followed by the addition of 500Pg/ml of MTT (Sigma). The blue crystals were dissolved in HCl/isopropanol and the absorbance was measured at 570 nm subtracting background at 690 nm using a Multiscan spectrophotometer (Labsystems, Finland).

Formation of Secondary Neurospheres To study the generation of secondary neurospheres, 500,000 newly passed NSCs were placed in each well of a six well plate, and treated with factors of interest. After 3 days, the total number of formed secondary neurospheres was counted.

Cell Differentiation NSCs were plated onto poly-DL-ornithine coated 48 well culture dishes (80,000 cells/well, Falcon, Sweden), and incubated for five days at 37qC in the presence of various factors. Cells were fixed using 4% PFA in PBS, and neurons, astro- cytes and oligodendrocytes were analyzed using immunocytochemistry.

Immunocytochemistry Cells were typically fixed in 4% PFA in PBS for 15-20 minutes at room tem- perature, washed in PBS and blocked for 30 minutes to one hour in 1-5% BSA, 0.1% TritonX-100 in PBS (PBS-T).

Non Fluorescent Staining BRCA-1, BrdU, CNPase, ERD, ERE, GFAP, Nestin and E-III-Tubulin

Endogenous peroxidases were inhibited with 0.3% H2O2 for 30 minutes at room temperature. For BrdU staining, the DNA was denatured in 2M HCl for 20 minutes to en- able the antibody to bind to the antigen. Primary antibodies were typically incu- bated over night in 0.1% TritonX-100/PBS. In order to get an increased signal, an amplification step was performed in which a secondary biotinylated antibody was added for 2 hours followed by Avidin-HRP complex for one hour, and visu- alization using 3,3´-diaminobenzidine (DAB) as a substrate.

35 Fluorescent Staining BrdU/BRCA-1, CNPase/GFAP, E-III-Tubulin/GFAP, ERE/AR, Nestin/MC3R and Nestin/MC4R Primary antibodies were typically incubated overnight followed by incubation with Cy2/Cy3 conjugated secondary antibodies for 30-40 minutes at room tem- perature. In the case of ERE/AR double staining, a secondary biotinylated anti chicken body antibody was added for ERE, followed by simultaneous incubation with avidin-FITC complex and secondary Cy2 conjugated anti rabbit antibody for AR. The BrdU/BRCA-1 double staining was performed sequentially with the complete BRCA-1 staining finalized before the DNA was denatured. The cells were then incubated with BrdU antibody overnight followed by secondary anti- body for one hour at room temperature. In the case of MC3R and MC4R double staining, cells were permeabilized for 5 minutes using 0.1% Triton-X in PBS. Blocking and antibodies were diluted in 2% BSA in PBS. MC3R and MC4R were visualized using secondary Cy3 and Cy2 conjugated antibodies, respectively. In some cases, nuclei were visualized using 4,6-Diamidine-2-phenylindole- dihydrochloride (DAPI, Sigma, 2Pg/ml).

Immunohistochemistry

BrdU Staining Paraffin embedded brains were sectioned, dewaxed in xylene, rehydrated in decreasing concentrations of ethanol and processed for antigen retrieval in 10mM citric acid pH 6.0. Endogenous peroxidases were inhibited in 3% H2O2 at 37qC for 10 minutes, and the DNA was denatured in 2M HCl for 30 minutes in 37qC, followed by neutralization in 0.05 M Borat buffer pH 8.5. Sections were treated with 0.1% trypsin in 37qC for 20 minutes and blocked for 20 minutes with 5% normal goat serum in diluent (1% BSA, 0.5% Tween-20 in PBS), fol- lowed by primary mouse monoclonal anti BrdU (Sigma, 1:500) in diluent over night at 4qC. Secondary biotinylated anti-mouse antibody was added in diluent for 2h, followed by Avidin-Horseradish peroxidase. Positive cells were visual- ized using DAB as a chromogen.

NR, BRCA-1 and MCR Staining To detect AR, ERs and BRCA-1 positive cells in rat brain, 13 Pm thick sections were cut on a cryostat (Leitz Digital, Germany), mounted onto Superfrost slides (Metzel-Gläser, Germany), and fixed for 10 min. Endogenous peroxidases were inhibited, sections were blocked using 5% goat serum in PBS-T, and incubated with primary antibodies overnight. Secondary biotinylated antibodies were

36 added followed by avidin-HRP incubation and visualization using DAB as a chromogen. To detect MC3R and MC4R in nestin positive cells of the rat brain, E17 par- affin sections were dewaxed, and processed for antigen retrieval with 10mM citric acid pH 6. Sections were blocked for 30 minutes with 5% BSA in PBS containing 0.05% TX-100, and incubated overnight at 4qC with nestin and MC3R or MC4R antibodies. Secondary Cy3 and Cy2 conjugated antibodies were added for 30 minutes at RT.

Stereology

Quantification of BrdU positive cells For quantification of the number of BrdU positive cells within the DG of the hippocampus, stereological principles were applied. The whole brain was cut in 10Pm sagital sections. A total of ten representative sections from the same area were then analyzed. The analyzed area was defined as the entire DG plus and an equally sized part of the adjacent CA4 such that the analyzed area was twice the thickness of DG containing also the sub granular cell layer. Using the CAST stereological program, the defined area was selected, and 30 sample frames were randomly chosen such that the SEM/mean<10% for each section. The same parameters were applied to all sections. Within these frames, the number of BrdU positive cells were counted using a Zeiss Axioskop100X objective and normalized to the area of the selected region. The criteria for a BrdU positive cell, was a nuclei completely filled with DAB staining or containing dark nu- clear spots with varying intensity. The mean number of cells per section, nor- malized by area, was obtained by pooling all counts from each section for each rat. The average number of BrdU positive cells/mm2 was plotted for the control and the treated animals. Student's t-test was used for statistical analysis.

37 Table 3. Antibodies Used. rb, rabbit; gt, goat; m, mouse; ch, chicken. WB, Western Blot- ting; ICC, Immunocytochemistry; IHC, Immunohistochemistry. Antibody Host Dilution Dilution Company Used in WB ICC/IHC Study Actin rb 1:2000 - Sigma I,II,III,IV AR rb 1:400 1:500 Santa Cruz III E-III-tubulin m 1:2000 1:500 Biosite I,III,IV BRCA-1 gt 1:200 1:100 Santa Cruz II BrdU* m - - Dako III BrdU m - 1:1000/1:200 Sigma I,II,III,IV ERD rb 1:1000 1:1000 Calbiochem I ERE ch 1:1500 1:1000 J.Å Gustafsson I CNPase m 1:2000 1:500 Sigma I,III,IV Cyclin D1 m 1:1000 - Santa Cruz III, IV EGFR rb 1:1000 - Santa Cruz IV p-EGFR† gt 1:500 - Santa Cruz IV GFAP m 1:2000 1:200 Sigma I,III,IV GFAP rb - 1:200 Sigma I,III,IV MC3R rb 1:500 1:200 Santa Cruz IV MC4R gt 1:500 1:200 Santa Cruz IV Nestin m - 1:200 Chemicon IV Nestin rb 1:1000 1:1000/1:500 Chemicon I,II,III,IV NeuN m - 1:200 Chemicon III p18 rb 1:500 - Santa Cruz III p21 m 1:500 - BD Biosciences I,III p27 m 1:2000 - BD Biosciences I,III p57 rb 1:500 - Santa Cruz I,III pRb m 1:1000 - BD Biosciences III,IV Secondary biotinylated ch/gt/r/m - 1:200 Vector Laborato- I,II,III,IV ries Avidin-Texas Red ch/gt/r/m - 1:100 Vector Laborato- I,II,III,IV ries Secondary Cy2/3 ch/gt/r/m - 1:300 Jackson Laborato- I,II,III,IV ries Secondary FITC** m Dako III *used for flow cytometry (1:100).** used for flow cytometry (1:200). † (Tyr1173)

38 Molecular Biology

RT-PCR The presence of transcripts for genes of interest in embryonic and adult NSCs was determined by reverse transcriptase polymerase chain reaction (RT-PCR) using total RNA isolated by using the Chomczynski and Sacchi method (Chomczynski and Sacchi, 1987). cDNA was made using reverse transcriptase (Invitrogen) according to the manufacture's instruction. The amplified PCR products were run on an agarose gel.

Table 4. Primers Used. F, Forward primer; R, reverse primer Gene Primer sequence Size (bp) Paper

-Actin F 5´-TTGGCGTACAGGTCTTTGCGG-3´ I,II,IV E 520 E-Actin R 5´-TGTTTGAGACCTTCAACACCC-3´ AR F 5´-GATATTACCTCCTGCTGCT-3´ III 496 AR R 5´-GAGCGTTCCAGAATCTGTT-3´ ER F 5´-GCCAGTCGAGCATCACTTACGGT-3´ I D 580 ERD R 5´-GTGCTTCAACATTCTCCCTCCTC-3´ ER pair1 F 5´-TTCCCGGCAGCACCAGTAACC-3´ I E 244 EREpair1 R 5´-TCCCTCTTTGCGTTGGACTA-3´ ER pair2 F 5´-GAGCTCAGCCTGTTGGACC-3´ I E 298 EREpair2 R 5´-GCCTTCACACAGAGATACTC-3´ MC3R F 5´-CGCCGATAACCATGAACTC-3´ IV 235 MC3R R 5´-AAGTACATGGGGGAGTGCAG-3´ MC4R F 5´-CTTTTACGCGCTCCAGTACC-3´ IV 163 MC4R R 5´-CAGCATGGTGAAGAACATGG-3´

39 AIMS OF THE PRESENT INVESTIGATION

The overall aim of this thesis was to investigate the role which hormones and hormone related regulators have on NSC proliferation and fate determination.

In particular:

1. To study the effect of the female sex hormone estrogen on prolifera- tion and fate determination in vitro of embryonic and adult NSCs iso- lated from the rat brain. 2. To study the effect of the synthetic testosterone analogue nandrolone on proliferation and fate determination in vitro and in vivo of embry- onic and adult NSCs isolated from the rat brain. 3. To characterize the BRCA-1 expression in the developing brain, with emphasis on regions containing proliferating precursor cells. In addi- tion, study the mRNA and protein expression of BRCA-1 in cultured NSCs. 4. To study the effect of the melanocortin D-MSH, on proliferation in vi- tro of embryonic NSCs isolated from the rat brain.

40 RESULTS AND DISCUSSION

Estrogen Affects the Proliferation and Fate Determination of NSCs (PaperI) Estrogen has many important functions in the nervous system where it for ex- ample promotes growth and survival of neurons. Whether the precursors of the neural cells, the NSCs, would also be regulated by estrogen was previously un- known. In this study we showed that the receptors for estrogen are expressed in the ventricular wall of the embryonic and adult rat brain, which are areas containing proliferating precursors. Also, ERs were expressed by the cultured embryonic and adult NSCs. Estrogen reduced the proliferation of EGF stimulated NSCs through involvement of the ERs, as shown by using the ERD and ERE antago- nist ICI 182 780. This decreased proliferation was in embryonic NSCs associ- ated with an up-regulation in the cdk inhibitor p21. In addition to affecting NSC proliferation, estrogen also affected NSC fate determination in vitro, by increas- ing the fraction of neurons to glial cells. This effect on differentiation was only seen in embryonic NSCs, suggesting that embryonic and adult NSCs have a different responsiveness to the hormone.

BRCA-1 is localized to Proliferating Neural Precursors and Down Regulated upon Differentiation (Paper II) The tumor suppressor BRCA-1 is known for regulating proliferation both during development and in estrogen responsive breast and ovarian cancer cells. Little is known about the role of BRCA-1 is the brain. In light of that estrogen regulates proliferation of NSCs; we were interested if the proliferating precursors in the brain expressed BRCA-1. Using in situ hybridization, immunohisto/cyto-chemistry, and western blot- ting we showed that BRCA-1 is expressed in the neuroepithelium containing neuronal precursor cells, as well as in cultured proliferating adult and embryonic NSCs. The expression of BRCA-1 decreased during development, but remained in proliferating precursors in the postnatal cerebellum, as well as in the ventricu-

41 lar lining of the adult rat brain. By differentiating these NSCs in vitro, we have observed a decreased expression of BRCA-1, suggesting a role of BRCA-1 in the regulation of NSC proliferation.

19-Nortestosterone Influences Neural Stem Cell Proliferation and Neurogenesis in the Rat Brain (PaperIII) We have previously shown that that the female sex hormone, estrogen, affects NSC proliferation and fate determination. We were therefore interested to study if the male sex hormone analogue, 19-nortestosterone, commonly referred to as nandrolone, would also influence the proliferation and fate determination of NSCs. In this study we showed that the androgen receptor is expressed both in areas of the embryonic and adult brain containing proliferating precursors, and in cultured NSCs. Nandrolone decreased the proliferation of EGF stimulated em- bryonic and adult NSCs through involvement of the AR. However, nandrolone did not affect the protein levels of any cdk inhibitors belonging to the Cip/Kip family. In addition to affecting NSC proliferation, nandrolone increased the neuronal fate of embryonic NSC in vitro. In light of our in vitro findings, we were interested if nandrolone would affect NSCs in vivo. After five days of nandrolone injections a reduction in the number of BrdU positive cells in the DG of the adult hippocampus in male, female and pregnant female rats by 30%, 22% and 36% respectively were observed. Inter- estingly, nandrolone does not only affect NSC proliferation in vivo, the effect varies with the sex and the hormonal status of the rat. This study shows that nandrolone affects NSC proliferation both in vitro and in vivo, and influences embryonic neurogenesis in vitro.

D-MSH is a Mitogen for Embryonic NSCs (Paper IV) Melanocortins regulate many physiological functions in the body. If D-MSH would also regulate functions in NSCs was previously unknown. In this paper we have studied the effect of D-MSH on proliferation of embry- onic rat NSCs. mRNA and protein expression data show that the MC3R and MC4R are present in the developing neuroepithelium, and in cultured NSCs. D-MSH doubled the proliferation of EGF starved NSCs, suggesting that D- MSH can counteract the cell cycle exit upon EGF withdrawal. This induced proliferation was concentration dependent, and probably mediated through MC4R, as shown by using the receptor antagonist HS014. Protein levels were up-regulated of cyclin D1 and pRb in response to D-MSH treatment, showing effects on cell cycle regulators, which explains the increased proliferation.

42 In addition, D-MSH increased the formation of secondary neurospheres, indi- cating that D-MSH stimulates the self renewal capacity of NSCs. This study shows that D-MSH plays an important role in regulating NSC pro- liferation and turn over by affecting important cell cycle regulators, identifying D-MSH a novel NSC.

43 GENERAL DISCUSSION AND FUTURE PERSPECTIVE

In the nervous system, sex hormones such as estrogen make up an important group of factors that affects growth, survival, maintenance of neurons and con- tributes to synaptic plasticity. Since NSCs are precursors for neurons, we hy- pothesized that estrogen might have important functions also in NSCs. In paper I and III, we have studied the effects of female sex hormone estro- gen and the anabolic steroid nandrolone, on NSC proliferation and fate determi- nation. Both estrogen and nandrolone decreased the proliferation of NSCs, and increased the neuronal fate of embryonic cells. In the case of estrogen, the de- crease in NSC proliferation was mediated through the inhibitor of cyclin de- pendent kinases, p21, indicating that estrogen can regulate the self renewal ca- pacity by affecting the NSC cell cycle machinery. In light of the increasing problem with abuse of anabolic steroids, we wondered if nandrolone would also have affects on NSC turn over in vivo. Doses comparable with heavy abuse of nandrolone decreased the number of BrdU positive cells in the DG, a region in hippocampus containing NSCs, by 30%, 22%, and 36% in male, female, and pregnant female rats. While estrogen is known to regulate levels of both cell cycle proteins and growth factors, the estrogen receptors are in turn regulated by for example tran- scription factors such as BRCA-1. This protein is important both during em- bryogenesis and in tumerogenesis in estrogen responsive cancer cells. In paper II, we identified BRCA-1 as a potential marker for NSCs, as BRCA-1 mRNA and protein are expressed by proliferating precursors in the embryonic and adult rodent brain, as well as in cultured NSCs. Upon differentiation, the BRCA-1 expression was diminished, and in post mitotic neurons and glial cells derived from NSCs, BRCA-1 is hardly detectable. Usually increased cAMP levels stimulate cell growth. We were interested if the melanocortin, D-MSH, would affect NSC proliferation. D-MSH increase cAMP levels in cells through involvement of their Gs-protein coupled receptors located at the plasma membrane (Gantz and Fong, 2003). In paper IV we showed that the two most abundant brain subtypes of the melanocortin receptors, MC3R and MC4R, are expressed by cells in the devel- oping neuroepithelium, as well as by NSCs in culture. D-MSH increased NSC proliferation two-fold in a concentration dependent manner via involvement of MC4R, as shown by receptor antagonists. This induced proliferation was ac-

44 companied by increased levels of both cyclinD1 and pRb, explaining the effects on proliferation The identification of stem cells in the adult human brain has changed the view of the brain. Before the identification of adult human NSCs, the birth of new neurons in the fully developed brain was considered less likely. As the population is getting increasingly older, neurodegenerative disorder such as Alzheimer’s and Parkinson’s disease are becoming an increasing prob- lem, both out of an economical and a social point of view. At the present mo- ment, the treatment strategy involves drug therapy which only slows down the disease progression, but the neurons are lost for good. Stem cells isolated from the brain could in the future be used to replace these lost neurons. There are two main plausible ways to use neural stem cells for this type of therapy. First, injection of either NSCs or mature neurons derived from NSCs into the brain. In the case of injecting stem cells, the cell must be preprogrammed into forming the appropriate cell type, for instance dopaminergic or cholinergic neu- rons. One of the potential problems of injecting undifferentiated stem cells into the brain is the possibility of tumor formation. An alternative is injection of mature neurons which can be derived from NSCs. In either case it is crucial to understand how NSC proliferation and fate determination is regulated. Second, since NSCs are located in parts of the adult brain, these cells could be manipulated in situ to proliferate, migrate and form new neurons at the sites of neuronal loss. While this alternative would be less invasive, and could there- fore be advantageous, many problems must first be solved. Not is it only critical to find factors that would direct differentiation of the stem cells in an appropri- ate manner, but also make sure that the cells are post mitotic, thereby avoiding tumor formation. More research into factors that influence NSC proliferation and differentiation is therefore necessary. This thesis has focused on studying effects of hormones on NSC proliferation and fate determination. Factor such as lipophilic hormones, is an attractive group of factors for treat- ment strategies. Hormones freely diffuse across the plasma membrane, and bind to their receptors which are present in NSCs. While estrogen and nandrolone will increase neurogenesis in culture, after differentiation there is a mixture of neurons and glial cells. Our in vivo studies showed that nandrolone decreased the proliferation of NSCs in the DG by 30% in males, 22% in females and 36% in pregnant females, showing that the effect of nandrolone depends on other sex hormones. It is therefore likely that sex hormones will work in conjunction with other factors which influence neurogenesis. The estrogen and androgen receptors are nuclear receptors which are tran- scription factors, affecting gene expression of a large number of target genes during development and in adult life. It is known that the estrogen receptors are not just regulating other genes, but is also subject of intrinsic regulation. For example, it is shown that BRCA-1 will inhibit ERD signaling in human breast and prostate cancer cells (Fan et al., 1999).

45 We have recently identified BRCA-1 as a possible regulator of NSC prolif- eration, and it would therefore be interesting to investigate whether estrogen signaling in NSCs is regulated by BRCA-1. This could be done in wild type cells, and then compared with effects seen in NSCs prepared from BRCA-1 mutated and ER null mice. Also, since there are species differences, it is impor- tant to study NSCs from the human brain to find out if the mechanisms observed in rodent also are true in the human case. Estrogen is an important hormone which has positive effects on the brain and bone density. While the positive effects on brain function in humans are de- bated, HRT clearly increase the incidence of breast cancer (Beral, 2003). As a result it is important to perform basic science where underlying mechanisms can be studied. In conclusion, in this thesis the effects of hormones and hormonal regulators have been studied. While we have found that sex hormones have profound ef- fects on NSCs both in vitro and in vivo, more studies are necessary before NSCs can have a place in human therapy.

46 ACKNOWLEDGEMENT

The research included in this thesis was carried out at the Department of Neuro- science, Unit for Neurobiology. There are numerous people that I would like to thank.

First and foremost: My supervisor, Professor Dan Lindholm for his constant flow of ideas, his posi- tive attitude and enthusiasm.

The neighboring professors Ted Ebendal and Finn Hallböök for encouragement.

Professor Håkan Aldskogius and Elena Kozlova for inspiring discussion and for making sure that one thing will always be true. Falukorv, that’s what’s for lunch!!

Co-authors, past and present members of the Dan Lindholm laboratory. In par- ticular:

The two fantastic post docs:

Beat, for always finding time to help me with whatever comes up. I am very, very, very grateful for all the help. The Miami shopping vacation, alligator site seeing and a trip to the nudist beach.

Rodrigo (some of you might know him as Rodriguez) for your special Spanish humor. For all the help especially with molecular biology problems, but also for interesting both scientific and “private” discussions. And of course for letting me drink the excellent wine that you bring all the way from Spain. Isabel for not only being the most talkable person in Sweden, but also having something in- teresting to tell.

Partypinglan Hanna, and Håkan (our own KennyG), for fun laughs in the office. Good luck with your own thesis.

Inga for help with various things.

Helena, fellow office mate.

47 Lotta for spreading a positive atmosphere in the lunch room and always taking time to discuss small and large issues! And for all the fun we had at Neurosci- ence Members of the rest of the Ebendal group: Sanna, Mitya and Annika, for inter- esting discussions during coffee. Fun dissertation parties and the pancake dinner at Mitya's.

The Hallböök group: Ullis for always being happy and positive, PH for interesting discussions, and Robert and Sojeong.

The Åkerman group: Karl Åkerman, Marie, Sylwia, “docenten” Jyrki, and Tomas for many fun mo- ments, interesting theories, and good laughs.

Marianne Jonsson for help with practical issues, and valuable discussions.

Absolut perfekta och bästa vännen Katti för allt stöd och för så många roliga stunder, och ibland lite tennis också. Tack, Tack, Tack. Johan för alla kul teorier om hur människan egentligen fungerar. Humor i världsklass!

Elle, precis som jag lite gnällig. För att du helt enkelt är dig själv, vilket är allde- les utmärkt!

Cissi, för att du är en riktigt god vän som alltid har tid för en pratstund, och ibland en liten springtur.

Kyle, för många fina år tillsammans. Allt stöd under hela min utbildning, och för att du trots allt tycker om mig......

Mamsen och pappsen: världens bästa föräldrar.

Anders, min favoritbrorsa.

48 REFERENCES

Alberg, AJ., Visvanathan, K., Helzlsouer, KJ. (1998). Epidemiology, prevention, and early detection of breast cancer. Curr Opin Oncol. 10(6):492-7. Altman J. (1969). Autoradiographic and histological studies of postnatal neurogenesis. IV. Cell proliferation and migration in the anterior forebrain, with special reference to persisting neurogenesis in the olfactory bulb. J Comp Neurol 137: 433-458. Altman, J., Das, GD. (1965). Autoradiographic and histological evidence of postnatal hippocampal neurogenesis in rats. J Comp Neurol 124:319-336. Alvarez-Buylla, A., Lim, DA. (2004). For the long run: maintaining germinal niches in the adult brain. Neuron. 41(5):683-6. Anderson DJ. (2001). Stem cells and pattern formation in the nervous system: the possi- ble versus the actual. Neuron. 30(1):19-35. Aprelikova, ON., Fang, BS., Meissner, EG., Cotter, S., Campbell, M., Kuthiala, A., Bessho, M., Jensen, RA., Liu, ET. (1999). BRCA-1-associated growth arrest is RB- dependent. Proc Natl Acad Sci U S A 96(21):11866-71. Arriza, JL., Fairman, WA., Wadiche, JI., Murdoch, GH., Kavanaugh, MP., Amara, SG. (1994). Functional comparison of three glutamate transporter subtypes cloned from human motor cortex. J. Neurosci. 14:5559-5569. Azcoitia, I., Sierra, A., Garcia-Segura, LM. (1999). Localization of -immunoreactivity in astrocytes of the adult rat brain. Glia. 26(3):260-7. Benjannet, S., Rondeau, N., Day, R., Chretien, M., Seidah, NG. (1991). PC1 and PC2 are proprotein convertases capable of cleaving proopiomelanocor-tin at distinct pairs of basic residues. Proc Natl Acad Sci U S A 88(9):3564-8. Beral V. (2003) Breast cancer and hormone-replacement therapy in the Million Women Study. Lancet. 362(9382):419-27. Erratum in: Lancet. 362(9390):1160. Beyer, C., Pawlak, J., Karolczak, M. (2003). Membrane receptors for oestrogen in the brain. J Neurochem. 87(3):545-50. Beyer, C., Raab, H. (1998). Nongenomic effects of oestrogen: embryonic mouse mid- brain neurones respond with a rapid release of calcium from intracellular stores. Eur J Neurosci. 10(1):255-62. Blakely, RD., Berson, HE., Fremeau, RT. Jr., Caron, MG., Peek, MM., Prince, HK., Bradley, CC. (1991).Cloning and expression of a functional serotonin transporter from rat brain. Nature. 354(6348):66-70. Bocchinfuso, WP., Korach, KS. (1997). Mammary gland development and tumorigenesis in estrogen receptor knockout mice. J Mammary Gland Biol Neoplasia. 2(4):323-34. Bonni, A., Sun, Y., Nadal-Vicens, M., Bhatt, A., Frank, DA., Rozovsky, I., Stahl, N., Yancopoulos, GD., Greenberg, ME. (1997). Regulation of Gliogenesis in the Central Nervous System by the JAK-STAT Signaling Pathway. Science. 278:477-483. Bork, P., Hofmann, K., Bucher, P., Neuwald, AF., Altschul, SF., Koonin, EV. (1997). A superfamily of conserved domains in DNA damage-responsive cell cycle checkpoint proteins. FASEB J. 11(1):68-76. Brazelton, TR., Rossi, FM., Keshet, GI., Blau, HM. (2000). From marrow to brain: ex- pression of neuronal phenotypes in adult mice. Science. 290(5497):1775-9.

49 Brodie, SG., Deng, CX. (2001) BRCA-1-associated tumorigenesis: what have we learned from knockout mice? Trends Genet. 17(10):18-22. Caccamo, D., Katsetos, CD., Herman, MM., Frankfurter, A., Collins, VP., Rubinstein, LJ. (1989). Immunohistochemistry of a spontaneous murine ovarian teratoma with neuroepithelial differentiation. Neuron-associated beta-tubulin as a marker for primi- tive neuroepithelium. Lab Invest. 60(3):390-8. Callebaut, I., Mornon, JP. (1997). From BRCA-1 to RAP1: a widespread BRCT module closely associated with DNA repair. FEBS Lett. 400(1):25-30. Carrer, HF., Araque, A., Buno, W. (2003). Estradiol regulates the slow Ca2+-activated K+ current in hippocampal pyramidal neurons. J Neurosci. 23(15):6338-44. Carroll, RS., Zhang, J., Black, PM. (1999). Expression of estrogen receptors alpha and beta in human meningiomas. J Neurooncol. 42(2):109-16. Chai, YL., Cui, J., Shao, N., Shyam, E., Reddy, P., Rao, VN. (1999). The second BRCT domain of BRCA-1 proteins interacts with p53 and stimulates transcription from the p21WAF1/CIP1 promoter. Oncogene. 18(1):263-8. Chan, FKM., Zhang, J., Chen, L., Shapiro, DN., Winoto, A. (1995). Identification of human/mouse p19, a novel CDK4/CDK6 inhibitor with homology to p16ink4. Mol. Cell. Biol. 15:2682-2688. Chen, J., Jackson, PK., Kirschner, MW., Dutta, A. (1995). Separate domains of p21 involved in the inhibition of CDK kinase and PCNA. Nature 374: 386-388. Chen, J., Silver, DP., Walpita, D., Cantor, SB., Gazdar, AF., Tomlinson, G., Couch, FJ., Weber, BL., Ashley, T., Livingston, DM., Scully, R. (1998). Stable interaction be- tween the products of the BRCA-1 and BRCA2 tumor suppressor genes in mitotic and meiotic cells. Mol Cell. 2(3):317-28. Chen, Y., Farmer, AA., Chen, CF., Jones, DC., Chen, PL., Lee, WH.(1996). BRCA-1 is a 220-kDa nuclear phosphoprotein that is expressed and phosphorylated in a cell cy- cle-dependent manner. Cancer Res. 56(14):3168-72. Chenn, A., McConnell, SK. (1995) Cleavage orientation and the asymmetric inheritance of Notch-1 immunoreactivity in mammalian neurogenesis. Cell 82: 631–641. Chiasson, BJ., Tropepe, V., Morshead, CM., van der Kooy, D. (1999). Adult mammalian forebrain ependymal and subependymal cells demonstrate proliferative potential, but only subependymal cells have neural stem cell characteristics. J Neurosci. 19(11):4462-71. Chomczynski, P., Sacchi, N. (1987). Single-Step Method of RNA isolation by Acid Guanidinium Thiocyanate-Phenol-Chloroform Extraction. Anal Biochem. 162:156- 159. Clarke, DL., Johansson, CB., Wilbertz, J., Veress, B., Nilsson, E., Karlstrom, H., Len- dahl, U., Frisen, J. (2000). Generalized potential of adult neural stem cells. Science. 288(5471):1660-3. Couse, JF., Bunch, DO., Lindzey, J., Schomberg, DW., Korach, KS. (1999). Prevention of the polycystic ovarian phenotype and characterization of ovulatory capacity in the estrogen receptor-alpha knockout mouse. Endocrinology. 140(12):5855-65. Couse, JF., Korach, KS. (1999). Estrogen receptor null mice: what have we learned and where will they lead us? Endocr Rev. 20(3):358-417. Erratum in: Endocr Rev. 20(4):459. De Wied, D., Croiset, G. (1991). Stress modulation of learning and memory processes. Methods Achiev Exp Pathol 15:167-99. Debus, E., Weber, K., Osborn, M. (1983). Monoclonal antibodies specific for glial fibril- lary acidic (GFA) protein and for each of the neurofilament triplet polypeptides. Dif- ferentiation. 25(2):193-203.

50 Deslypere, JP., Young, M., Wilson, JD., McPhaul, MJ. (1992). Testosterone and 5 alpha- dihydrotestosterone interact differently with the androgen receptor to enhance tran- scription of the MMTV-CAT reporter gene. Mol Cell Endocrinol. 88(1-3):15-22. Doetsch, F., Caille, I., Lim, DA., Garcia-Verdugo, JM., Alvarez-Buylla, A. (1999). Sub- ventricular zone astrocytes are neural stem cells in the adult mammalian brain. Cell. 97(6):703-16. Doisneau-Sixou, SF., Sergio, CM., Carroll, JS., Hui, R., Musgrove, EA., Sutherland, RL. (2003). Estrogen and antiestrogen regulation of cell cycle progression in breast can- cer cells. Endocr Relat Cancer. 10(2):179-86. Dulic, V., Kaufmann, WK., Wilson, SJ., Tlsty, TD., Lees, E., Harper, JW., Elledge, SJ., Reed, SI. (1994). p53-Dependent inhibition of cyclin-dependent kinase activities in human fibroblasts during radiation-induced G1 arrest. Cell 76:1013-1023. El-Deiry, WS., Tokino, T., Velculescu, VE., Levy, DB., Parsons, R., Trent, JM., Lin, D., Mercer, WE., Kinzler, KW., Vogelstein, B. (1993). WAF1, a potential mediator of p53 tumor suppression. Cell 75:817-825. Emsley, JG., Arlotta, P., Macklis, JD. (2004). Star-cross'd neurons: astroglial effects on neural repair in the adult mammalian CNS. Trends Neurosci. 27(5):238-40. Eng, LF., Ghirnikar, RS., Lee, YL. (2000). Glial fibrillary acidic protein: GFAP-thirty- one years (1969-2000). Neurochem Res. 25(9-10):1439-51. Eriksson, PS., Perfilieva, E., Bjork-Eriksson, T., Alborn, AM., Nordborg, C., Peterson, DA., Gage, FH. (1998). Neurogenesis in the adult human hippocampus. Nat Med 4:1313-1307. Erlandsson, A., Enarsson, M., Forsberg-Nilsson, K. (2001). Immature Neurons From CNS Stem Cells Proliferate in Respons to Platelet-derived Growth Factor. J Neuro- sci. 21(13):3483-3491. Fan, S., Ma, YX., Wang, C., Yuan, RQ., Meng, Q., Wang, JA., Erdos, M., Goldberg, ID., Webb, P., Kushner, PJ., Pestell, RG., Rosen, EM. (2001). Role of direct interaction in BRCA-1 inhibition of estrogen receptor activity. Oncogene 20(1):77-87. Fan, S., Wang, J., Yuan, R., Ma, Y., Meng, Q., Erdos, MR., Pestell, RG., Yuan, F., Auborn, KJ., Goldberg, ID., Rosen, EM. (1999). BRCA-1 inhibition of estrogen re- ceptor signaling in transfected cells. Science. 284(5418):1354-6. Fan, W., Boston, BA., Kesterson, RA., Hruby, VJ., Cone, RD. (1997). Role of melano- cortinergic neurons in feeding and the agouti obesity syndrome. Nature 385(6612):165-8. Feng, JD., Dao, T., Lipton, JM. (1987). Effects of preoptic microinjections of alpha- MSH on fever and normal temperature control in rabbits. Brain Res Bull. 18(4):473- 7. Ferrari, G., Cusella-De Angelis, G., Coletta, M., Paolucci, E., Stornaiuolo, A., Cossu, G., Mavilio, F. (1998). Muscle regeneration by bone marrow-derived myogenic pro- genitors. Science. 279(5356):1528-30. Erratum in: Science 1998 281(5379):923. Ferreira, A., Caceres, A. (1991). Estrogen-enhanced neurite growth: evidence for a selec- tive induction of Tau and stable microtubules. J Neurosci. 11(2):392-400. Finley, SK., Kritzer, MF. (1999). Immunoreactivity for intracellular androgen receptors in identified subpopulations of neurons, astrocytes and oligodendrocytes in primate prefrontal cortex. J Neurobiol. 40(4):446-57. Fisher, CR., Graves, KH., Parlow, AF., Simpson, ER. (1998). Characterization of mice deficient in aromatase (ArKO) because of targeted disruption of the cyp19 gene. Proc Natl Acad Sci U S A. 95(12):6965-70. Furukawa, T., Mukherjee, S., Bao, ZZ., Morrow, EM., Cepko, CL. (2000). rax, Hes1, and notch1 promote the formation of Muller glia by postnatal retinal progenitor cells. Neuron. 26(2):383-94.

51 Gaiano N, Nye JS, Fishell G. (2000). Radial glial identity is promoted by Notch1 signal- ing in the murine forebrain. Neuron. 26(2):395-404. Galli, R., Borello, U., Gritti, A., Minasi, MG., Bjornson, C., Coletta, M., Mora, M., De Angelis, MG., Fiocco, R., Cossu, G., Vescovi, AL. (2000). Skeletal myogenic po- tential of human and mouse neural stem cells. Nat Neurosci. 3(10):986-91. Gantz, I., Fong, TM. (2003). The melanocortin system.Am J Physiol Endocrinol Metab. 284(3):468-74. Gantz, I., Konda, Y., Tashiro, T., Shimoto, Y., Miwa, H., Munzert, G., Watson, SJ., DelValle, J., Yamada, T (1993). Molecular cloning of a novel melanocortin receptor. J Biol Chem 268(11):8246-50. Garcia-Verdugo, JM., Doetsch, F., Wichterle, H., Lim, DA., Alvarez-Buylla, A. (1998). Architecture and cell types of the adult subventricular zone: in search of the stem cells. J Neurobiol. 36(2):234-48. Garner, CC., Brugg, B., Matus, A. (1988). A 70-kilodalton microtubule-associated pro- tein (MAP2c), related to MAP2. J Neurochem. 50(2):609-15. Gould, E., Woolley, CS., Frankfurt, M., McEwen, BS. (1990). Gonadal steroids regulate dendritic spine density in hippocampal pyramidal cells in adulthood. J Neurosci. 10(4):1286-91. Gould, E., Vail, N., Wagers, M., Gross, CG. (2001). Adult-generated hippocampal and neocortical neurons in macaques have a transient existence. Proc Natl Acad Sci U S A 98:10910-10917. Green, S., Walter, P., Kumar, V., Krust, A., Bornert, JM., Argos, P., Chambon, P. (1986). Human oestrogen receptor cDNA: sequence, expression and homology to v- erb-A. Nature. 320(6058):134-9. Greene, GL., Gilna, P., Waterfield, M., Baker, A., Hort, Y., Shine, J. (1986). Sequence and expression of human estrogen receptor complementary DNA. Science. 231(4742):1150-4. Gritti, A., Frolichsthal-Schoeller, P., Galli, R., Parati, EA., Cova, L., Pagano, SF., Bjorn- son, CR., Vescovi, AL. (1999). Epidermal and fibroblast growth factors behave as mitogenic regulators for a single multipotent stem cell-like population from the sub- ventricular region of the adult mouse forebrain. J Neurosci. 19(9):3287-97. Gross RE, Mehler MF, Mabie PC, Zang Z, Santschi L, Kessler JA. (1996). Bone morphogenetic proteins promote astroglial lineage commitment by mammalian sub- ventricular zone progenitor cells. Neuron. 17(4):595-606. Gu, Q., Korach, KS., Moss, RL. (1999). Rapid action of 17beta-estradiol on kainate- induced currents in hippocampal neurons lacking intracellular estrogen receptors. Endocrinology. 140(2):660-6. Gu, Y., Turek, CW., Morgan, DO. (1993). Inhibition of CDK2 activity in vivo by an associated 20K regulatory subunit. Nature 366: 707-710. Guan, KL., Jenkins, CW., Li, Y., Nichols, MA., Wu, X., O'Keefe, CL:, Matera, AG., Xiong, Y. (1994). Growth suppression by p18, a p16INK4/MTS1 and p15INK4b/MTS2 re- lated CDK6 inhibitor, correlates with wild-type pRb function. Genes &Dev. 8:2939- 2952. Gundlah, C., Kohama, SG., Mirkes, SJ., Garyfallou, VT., Urbanski, HF., Bethea, CL. (2000). Distribution of estrogen receptor beta (ERbeta) mRNA in hypothalamus, midbrain and temporal lobe of spayed macaque: continued expression with hormone replacement. Brain Res Mol Brain Res. 76(2):191-204. Haigh, JR., Norenberg, K., Parsons, SM. (1994). Acetylcholine active transport by rat brain synaptic vesicles. Neuroreport. 5:773-776. Hakem, R., de la Pompa, JL., Sirard, C., Mo, R., Woo, M., Hakem, A., Wakeham, A., Potter, J., Reitmair, A., Billia, F., Firpo, E., Hui, CC., Roberts, J., Rossant, J., Mak,

52 TW. (1996). The tumor suppressor gene BRCA-1 is required for embryonic cellular proliferation in the mouse. Cell. 85(7):1009-23. Hakem, R., de la Pompa, JL., Mak, TW. (1998). Developmental studies of BRCA-1 and Brca2 knock-out mice. J Mammary Gland Biol Neoplasia. 3(4):431-45. Hannon, GJ. Beach, D. (1994). p15INK4b is a potential effector of TGF -induced cell cycle arrest. Nature 371: 257-261. Hanstein, B., Eckner, R., DiRenzo, J., Halachmi, S., Liu, H., Searcy, B., Kurokawa, R., Brown, M.(1996). p300 is a component of an estrogen receptor coactivator complex. Proc Natl Acad Sci U S A. 93(21):11540-5. Hardin JA, Brockway PD, Gall DG. (1998). Atrial natriuretic peptide-induced secretory responses in rabbit vs rat ileum. Comp Biochem Physiol A Mol Integr Physiol. 19(3):833-8. Harper, JW., Adami, GR., Wei, N., Keyomarsi, K., Elledge, SJ. (1993). The p21 CDK- interacting protein Cip1 is a potent inhibitor of G1 cyclin-dependent kinases. Cell 75: 805-816. Hartman, BK., Agrawal, HC., Agrawal, D., Kalmbach, S.(1982). Development and maturation of central nervous system myelin: comparison of immunohistochemical localization of proteolipid protein and basic protein in myelin and oligodendrocytes. Proc Natl Acad Sci U S A. 79(13):4217-20. Heinlein, CA., Chang, C. (2002a). Androgen Receptor (AR) Coregulators: An Overview. Endocr Rev. 23(2):175-200. Heinlein, CA., Chang, C. (2002b). The roles of androgen receptors and androgen-binding proteins in nongenomic androgen actions. Mol Endocrinol. 16(10):2181-7. Hirai, H., Roussel, MF., Kato, J., Ashmun, RA., Sherr, CJ. (1995). Novel INK4 proteins, p19 and p18, are specific inhibitors of the cyclin D-dependent kinases CDK4 and CDK6. Mol. Cell. Biol. 15:2672-2681. Hirano, M., Goldman, JE. (1988). Gliogenesis in rat spinal cord: evidence for origin of astrocytes and oligodendrocytes from radial precursors. J Neurosci Res. 21(2- 4):155-67. Horsburgh, K., Macrae, IM., Carswell, H.(2002). Estrogen is neuroprotective via an apolipoprotein E-dependent mechanism in a mouse model of global ischemia. J Cereb Blood Flow Metab. 22(10):1189-95. Hösli, E., Jurasin, K., Ruhl, W., Luthy, R., Hosli, L. (2001). Colocalization of androgen, estrogen and cholinergic receptors on cultured astrocytes of rat central nervous sys- tem. Int J Dev Neurosci. 19(1):11-9. Houle, J., Fedoroff, S. (1983). Temporal relationship between the appearance of vimentin and neural tube development.Brain Res. 285(2):189-95. Hu, E., Tontonoz, P., Spiegelman, BM. (1995). Transdifferentiation of myoblasts by the adipogenic transcription factors PPAR gamma and C/EBP alpha. Proc Natl Acad Sci U S A. 92(21):9856-60. Hunt, T.(1991). Cyclins and their partners: from a simple idea to complicated reality. Semin Cell Biol. 2(4):213-22. Johansson, CB., Momma, S., Clarke, DL., Risling, M., Lendahl, U. Frisen, J. (1999). Identification of a neural stem cell in the adult mammalian central nervous system. Cell 96:25-34. Johansson-Steensland, P., Nyberg, F., Chahl, L. (2002). The anabolic androgenic steroid, 19-nortestosterone decanoate, increases the density of Fos-like immunoreactive neu- rons in limbic regions of guinea-pig brain. Eur J Neurosci.15(3):539-44. Johe, KK., Hazel, TG., Muller, T., Dugich-Djordjevic, MM., McKay, RD. (1996). Single factors direct the differentiation of stem cells from the fetal and adult central nervous system. Genes Dev 10:3129–3140.

53 Kalyani, A., Hobson, K., Rao, MS. (1997). Neuroepithelial stem cells from the embry- onic spinal cord: isolation, characterization, and clonal analysis. Dev Biol. 186(2):202-23. Kalyani, AJ., Piper, D., Mujtaba, T., Lucero, MT., Rao, MS. (1998). Spinal cord neu- ronal precursors generate multiple neuronal phenotypes in culture. J Neurosci. 18(19):7856-68. Kawai, H., Li, H., Chun, P., Avraham, S., Avraham, HK. (2002). Direct interaction be- tween BRCA-1 and the estrogen receptor regulates vascular endothelial growth fac- tor (VEGF) transcription and secretion in breast cancer cells. Oncogene. 21(50):7730-9. Kerr, P., Ashworth, A. (2001). New complexities for BRCA-1 and BRCA2.Curr Biol 11(16):R668-76. Kindlundh, AM., Lindblom, J., Bergstrom, L., Wikberg, JE., Nyberg, F. (2001). The anabolic-androgenic steroid 19-nortestosterone decanoate affects the density of do- pamine receptors in the male rat brain. Eur J Neurosci. 13(2):291-6. Kindlundh, AM., Lindblom, J., Bergstrom, L., Nyberg, F. (2003). The anabolic- androgenic steroid nandrolone induces alterations in the density of serotonergic 5HT1B and 5HT2 receptors in the male rat brain. Neuroscience. 119(1):113-20. Kintner, C. (2002). Neurogenesis in embryos and in adult neural stem cells. J Neurosci. 22(3):639-43. Kistler-Heer, V., Schlumpf, M., Lichtensteiger, W. (1998). Melanocortin and MCH precursor-derived NEI effects on striatum-midbrain cocultures. Peptides 19(8):1317- 27. Koonin, EV., Altschul, SF., Bork, P. (1996). BRCA-1 protein products ... Functional motifs... Nat Genet. 13(3):266-8. Kopen, GC., Prockop, DJ., Phinney, DG. (1999). Marrow stromal cells migrate through- out forebrain and cerebellum, and they differentiate into astrocytes after injection into neonatal mouse brains. Proc Natl Acad Sci U S A. 96(19):10711-6. Kowalczyk, A., Filipkowski, RK., Rylski, M., Wilczynski, GM., Konopacki, FA., Jawor- ski, J., Ciemerych, MA., Sicinski, P., Kaczmarek L. (2004). The critical role of cy- clin D2 in adult neurogenesis. J Cell Biol 167(2):209-13. Krege, JH., Hodgin, JB., Couse, JF, Enmark, E., Warner, M., Mahler, JF., Sar, M., Korach, KS., Gustafsson, JA., Smithies, O. (1998). Generation and reproductive phenotypes of mice lacking estrogen receptor beta. Proc Natl Acad Sci U S A. 95(26):15677-82. Kuhlbrodt, K., Herbarth, B., Sock, E., Hermans-Borgmeyer, I., Wegner, M. (1998). Sox10, a novel transcriptional modulator in glial cells.J Neurosci. 18(1):237-50. Kuiper, GG., Enmark, E., Pelto-Huikko, M., Nilsson, S., Gustafsson, JA. (1996). Clon- ing of a novel receptor expressed in rat prostate and ovary. Proc Natl Acad Sci U S A. 93(12):5925-30. Lagasse, E., Connors, H., Al-Dhalimy, M., Reitsma, M., Dohse, M., Osborne, L., Wang, X., Finegold, M., Weissman, IL., Grompe, M. (2000). Purified hematopoietic stem cells can differentiate into hepatocytes in vivo. Nat Med. 6(11):1229-34. Lamb, TM., Harland, RM. (1995). Fibroblast growth factor is a direct neural inducer, which combined with noggin generates anterior-posterior neural pattern. Develop- ment 121:3627-3636. Lane, TF., Deng, C., Elson, A., Lyu, MS., Kozak, CA., Leder, P. (1995). Expression of BRCA-1 is associated with terminal differentiation of ectodermally and mesoder- mally derived tissues in mice. Genes Dev. 9(21):2712-22. Laywell, ED., Rakic, P., Kukekov, VG., Holland, EC., Steindler, DA. (2000). Identifica- tion of a multipotent astrocytic stem cell in the immature and adult mouse brain. Proc Natl Acad Sci U S A. 97(25):13883-8.

54 Le Greves, P., Huang, W., Johansson, P., Thornwall, M., Zhou, Q., Nyberg, F. (1997). Effects of an anabolic-androgenic steroid on the regulation of the NMDA receptor NR1, NR2A and NR2B subunit mRNAs in brain regions of the male rat. Neurosci Lett. 226(1):61-4. Lee, JC., Mayer-Proschel, M., Rao, MS. (2000). Gliogenesis in the central nervous sys- tem. Glia 30:105-121. Lehtimaki, T., Dastidar, P., Jokela, H., Koivula, T., Lehtinen, S., Ehnholm, C., Punno- nen, R. (2002). Effect of long-term hormone replacement therapy on atherosclerosis progression in postmenopausal women relates to functional apolipoprotein e geno- type. J Clin Endocrinol Metab. 87(9):4147-53. Lendahl, U., Zimmerman, LB., McKay, RD. (1990). CNS stem cells express a new class of intermediate filament protein. Cell. 60(4):585-95. Leranth, C., Roth, RH., Elsworth, JD., Naftolin, F., Horvath, TL., Redmond, DE Jr. (2000). Estrogen is essential for maintaining nigrostriatal dopamine neurons in pri- mates: implications for Parkinson's disease and memory. J Neurosci. 20(23):8604-9. Lewis RM, Wallace WC, Kanazir SD, Greengard P. (1983). Expression of cell-type- specific neuronal phosphoproteins. Cold Spring Harb Symp Quant Biol. 48 (1):347- 54. Li W, Cogswell CA, LoTurco JJ.(1998). Neuronal differentiation of precursors in the neocortical ventricular zone is triggered by BMP. J Neurosci. 18(21):8853-62. Lin, J., Reichner, C., Wu, X., Levine, AJ. (1996). Analysis of wild-type and mutant p21WAF-1 gene activities. Mol. Biol. Cell 16: 1786-1793. Lindblom, J., Schioth, HB., Larsson, A., Wikberg, JE., Bergstrom, L. (1998). Autoradio- graphic discrimi-nation of melanocortin receptors indicates that the MC3 subtype dominates in the medial rat brain. Brain Res 810(1-2):161-71. Lindqvist, AS., Johansson-Steensland, P., Nyberg, F., Fahlke, C. (2002). Anabolic an- drogenic steroid affects competitive behaviour, behavioural response to ethanol and brain serotonin levels. Behav. Brain Res 133(1):21-29. Liu, CY., Flesken-Nikitin, A., Li, S., Zeng, Y., Lee, WH. (1996). Inactivation of the mouse BRCA-1 gene leads to failure in the morphogenesis of the egg cylinder in early postimplantation development. Genes Dev. 10(14):1835-43. Liu, Y., Wu, Y., Lee, JC., Xue, H., Pevny, LH., Kaprielian, Z., Rao, MS. (2002). Oli- godendrocyte and astrocyte development in rodents: an in situ and immunohis- tological analysis during embryonic development. Glia. 40(1):25-43. Lorenzo, A., Diaz, H., Carrer, H., Caceres, A. (1992). Amygdala neurons in vitro: neurite growth and effects of estradiol. J Neurosci Res. 33(3):418-35. Lubahn, DB., Moyer, JS., Golding, TS., Couse, JF., Korach, KS., Smithies, O. (1993) Alteration of reproductive function but not prenatal sexual development after inser- tional disruption of the mouse estrogen receptor gene. Proc Natl Acad Sci U S A. 90(23):11162-6. Ludwig, T., Chapman, DL., Papaioannou, VE., Efstratiadis, A. (1997). Targeted muta- tions of breast cancer susceptibility gene homologs in mice: lethal phenotypes of BRCA-1, Brca2, BRCA-1/Brca2, BRCA-1/p53, and Brca2/p53 nullizygous em- bryos. Genes Dev. 11(10):1226-41. Luger, TA., Schwarz, T., Kalden, H., Scholzen, T., Schwarz, A., Brzoska, T. (1999). Role of epidermal cell-derived alpha-melanocyte stimulating hormone in ultraviolet light mediated local immunosuppression. Ann N Y Acad Sci. 885:209-16. Luine, VN. (1985). Estradiol increases choline acetyltransferase activity in specific basal forebrain nuclei and projection areas of female rats. Exp Neurol. 89(2):484-90. Lundkvist, J., Lendahl, U. (2001). Notch and the birth of glial cells. Trends Neurosci. 24(9):492-4.

55 Lynch, CS., Story, AJ. (2000). Dihydrotestosterone and estrogen regulation of rat brain androgen-receptor immunoreactivity. Physiol Behav. 69(4-5):445-53. Ma, Q., Kintner, C., Anderson, DJ. (1996). Identification of neurogenin, a vertebrate neuronal determination gene. Cell 87:43-52. MacLusky, NJ. (2004). Estrogen and Alzheimer's disease: the apolipoprotein connection. Endocrinology. 145(7):3062-4. MacLusky, NJ., Clark, AS., Naftolin, F., Goldman-Rakic, PS. (1987). Estrogen forma- tion in the mammalian brain: possible role of aromatase in sexual differentiation of the hippocampus and neocortex. Steroids. 50(4-6):459-74. Marquis, ST., Rajan, JV., Wynshaw-Boris, A., Xu, J., Yin, GY., Abel, KJ., Weber, BL., Chodosh, LA. (1995). The developmental pattern of BRCA-1 expression implies a role in differentiation of the breast and other tissues. Nat Genet. 11(1):17-26. Matsumoto, T., Honda, S., Harada, N. (2003). Neurological effects of aromatase defi- ciency in the mouse. J Steroid Biochem Mol Biol. 86(3-5):357-65. Matsuoka, S., Edwards, M., Bai, C., Parker, S., Zhang, P., Baldini, A., Harper, JW., Elledge, SJ. (1995). p57KIP2, a structurally distinct member of the p21CIP1 CDK in- hibitor family, is a candidate tumor suppressor gene. Genes & Dev. 9:650-662. Matus, A. (1988). Microtubule-associated proteins: their potential role in determining neuronal morphology. Annu Rev Neurosci. 11:29-44. McEwen, BS. (1983). Gonadal steroid influences on brain development and sexual dif- ferentiation. Int Rev Physiol. 27:99-145. McEwen, BS. (2001). Estrogens effects on the brain: multiple sites and molecular mechanisms. J. Appl. Physiol. 91:2785-2801. McGrew, LL., Lai, C J., Moon, RT. (1995). Specification of the antero-posterior neural axis through synergistic interaction of the Wnt signaling cascade with noggin and follistatin. Dev. Biol. 172, 337-342. McKay, R. (1997). Stem cells in the central nervous system. Science. 276(5309):66-71. Mehler, MF., Mabie, PC., Zhu, G., Gokhan, S., Kessler, JA. (2000). Developmental changes in progenitor cell responsiveness to bone morphogenetic proteins differen- tially modulate progressive CNS lineage fate. Dev Neurosci 22:74–85. Menard, CS., Harlan, RE. (1993). Up-regulation of androgen receptor immunoreactivity in the rat brain by androgenic-anabolic steroids. Brain Res. 622(1-2):226-36. Mermelstein, PG., Becker, JB., Surmeier, DJ. (1996). Estradiol reduces calcium currents in rat neostriatal neurons via a membrane receptor. J Neurosci. 16(2):595-604. Mezey, E., Chandross, KJ., Harta, G., Maki, RA., McKercher, SR. (2000). Turning blood into brain: cells bearing neuronal antigens generated in vivo from bone marrow. Sci- ence. 290(5497):1779-82. Miki, Y., Swensen, J., Shattuck-Eidens, D., Futreal, PA., Harshman, K., Tavtigian, S., Liu, Q., Cochran, C., Bennett, LM., Ding, W. (1994). A strong candidate for the breast and ovarian cancer susceptibility gene BRCA-1. Science 266(5182):66-71. Molne, M., Studer, L., Tabar, V., Ting, YT., Eiden, MV., McKay, RD. (2000). Early cortical precursors do not undergo LIF-mediated astrocytic differentiation. J Neuro- sci Res 59:301–311. Molofsky, AV., Pardal, R., Iwashita, T., Park, IK., Clarke, MF., Morrison, SJ. (2003). Bmi-1 dependence distinguishes neural stem cell self-renewal from progenitor pro- liferation. Nature. 425(6961):962-7. Morrison, SJ. (2001). Neuronal potential and lineage determination by neural stem cells. Curr Opin Cell Biol. 13(6):666-72. Mullen, RJ., Buck, CR., Smith, AM. (1992). NeuN, a neuronal specific nuclear protein in vertebrates. Development. 116(1):201-11. Mumm, JS., Kopan, R. (2000). Notch signaling: from the outside in. Dev Biol 228:151– 165.

56 Nadarajah, B., Parnavelas, JG.(2002). Modes of neuronal migration in the developing cerebral cortex. Nat Rev Neurosci. 3(6):423-32. Nagatsu, T., Levitt, M., Udenfriend, S. (1964). Tyrosine hydroxylase. The initial step in norepinephrine biosynthesis. J Biol Chem. 239:2910-7. Nakanishi, M., Robetorge, RS., Adami, GR., Pereira-Smith, OM., Smith, JR. (1995). Identification of the active region of the DNA synthesis inhibitory gene p21Sdi1/CIP1/WAF1. EMBO J. 14:555-563 Nakanishi, S., Inoue, A., Kita, T., Nakamura, M., Chang, AC., Cohen, SN., Numa, S. (1979). Nucleotide sequence of cloned cDNA for bovine corticotropinbetalipotropin precursor. Nature 278(5703):423-7. Nathan, BP., Jiang, Y., Wong, GK., Shen, F., Brewer, GJ., Struble, RG. (2002). Apoli- poprotein E4 inhibits, and apolipoprotein E3 promotes neurite outgrowth in cultured adult mouse cortical neurons through the low-density lipoprotein receptor-related protein. Brain Res. 928(1-2):96-105. Neish, AS., Anderson, SF., Schlegel, BP., Wei, W., Parvin, JD. (1998). Factors associ- ated with the mammalian RNA polymerase II holoenzyme. Nucleic Acids Res 26(3):847-53. Noda, A., Ning, Y., Venable, SF., Pereira-Smith, OM., Smith, JR. (1994). Cloning of senescent cell-derived inhibitors of DNA synthesis using an expression screen. Exp. Cell. Res. 211:90-98 Nurse, P. (2002). Cyclin dependent kinases and cell cycle control (nobel lecture). Chem- biochem. 3(7):596-603. Ogawa, S., Chester, AE., Hewitt, SC., Walker, VR., Gustafsson, JA., Smithies, O., Korach, KS., Pfaff, DW. (2000). Abolition of male sexual behaviors in mice lacking estrogen receptors alpha and beta (alpha beta ERKO). Proc Natl Acad Sci U S A 97(26):14737-41. Ohnuma, S., Harris, WA. (2003). Neurogenesis and the cell cycle. Neuron. 40(2):199- 208. Ohnuma, S., Philpott, A., Wang, K., Holt, CE., Harris, WA. (1999). p27Xic1, a Cdk inhibitor, promotes the determination of glial cells in Xenopus retina. Cell. 99(5):499-510. Ohnuma, S., Hopper, S., Wang, KC., Philpott, A., Harris, WA. (2002). Co-ordinating retinal histogenesis: early cell cycle exit enhances early cell fate determination in the Xenopus retina. Development. 129(10):2435-46. Orlic, D., Kajstura, J., Chimenti, S., Jakoniuk, I., Anderson, SM., Li, B., Pickel, J., McKay, R., Nadal-Ginard, B., Bodine, DM., Leri, A., Anversa, P. (2001). Bone mar- row cells regenerate infarcted myocardium. Nature 410(6829):701-5. Ouchi, T., Monteiro, AN., August, A., Aaronson, SA., Hanafusa, H. (1998). BRCA-1 regulates p53-dependent gene expression. Proc Natl Acad Sci U S A. 95(5):2302-6. Overturf, K., al-Dhalimy, M., Ou, CN., Finegold, M., Grompe, M.(1997). Serial trans- plantation reveals the stem-cell-like regenerative potential of adult mouse hepato- cytes. Am J Pathol. 151(5):1273-80. Paech, K., Webb, P., Kuiper, GG., Nilsson, S., Gustafsson, J., Kushner, PJ., Scanlan, TS. (1997). Differential ligand activation of estrogen receptors ERalpha and ERbeta at AP1 sites. Science. 277(5331):1508-10. Panchision, DM., McKay, RD. (2002). The control of neural stem cells by morphogenic signals. Curr Opin Genet Dev. 12(4):478-87. Papalopulu, N., Kintner, C. (1996). A posteriorising factor, retinoic acid, reveals that anteroposterior patterning controls the timing of neuronal differentiation in Xenopus neuroectoderm. Development 122:3409-3418 . Parnavelas JG, Nadarajah B. (2001). Radial glial cells. are they really glia? Neuron. 31(6):881-4.

57 Patrone, C., Ma, ZQ., Pollio, G., Agrati, P., Parker, MG., Maggi, A. (1996). Cross- coupling between insulin and estrogen receptor in human neuroblastoma cells. Mol Endocrinol. 10(5):499-507. Patrone, C., Pollio, G., Vegeto, E., Enmark, E., de Curtis, I., Gustafsson, J.A. Maggi, A. (2000). Estradiol induces differential neuronal phenotypes by activating estrogen re- ceptor alpha or beta. Endocrinology 141:1839-1845. Pevny, LH., Sockanathan, S., Placzek, M., Lovell-Badge, R. (1998). A role for SOX1 in neural determination. Development. 125(10):1967-78. Pike, CJ. (1999) Estrogen modulates neuronal Bcl-xL expression and beta-amyloid- induced apoptosis: relevance to Alzheimer's disease. J Neurochem 72(4):1552-63. Polyak, K., Kato, JY., Solomon, MJ., Sherr, CJ., Massagué, J., Roberts, JM., Koff, A. (1994). p27Kip1, a cyclin-CDK inhibitor, links transforming growth factor- and con- tact inhibition to cell cycle arrest. Genes & Dev. 8:9-22. Pritchard, LE., Turnbull, AV., White, A. (2002). Pro-opiomelanocortin processing in the hypothalamus: impact on melanocortin signalling and obesity. J Endocrinol. 172(3):411-21. Qi, Y., Cai, J., Wu, Y., Wu, R., Lee, J., Fu, H., Rao, M., Sussel, L., Rubenstein, J., Qiu, M. (2001). Control of oligodendrocyte differentiation by the Nkx2.2 homeodomain transcription factor. Development. 128(14):2723-33. Qiu, J., Bosch, MA., Tobias, SC., Grandy, DK., Scanlan, TS., Ronnekleiv, OK., Kelly, MJ. (2003). Rapid signaling of estrogen in hypothalamic neurons involves a novel G-protein-coupled estrogen receptor that activates protein kinase C. J Neurosci. 23(29):9529-40. Raballo, R., Rhee, J., Lyn-Cook, R., Leckman, JF., Schwartz, ML., Vaccarino, FM. (2000). Basic fibroblast growth factor (Fgf2) is necessary for cell proliferation and neurogenesis in the developing cerebral cortex. J Neurosci. 20(13):5012-23. Rajan, JV., Wang, M., Marquis, ST., Chodosh, LA. (1996). Brca2 is coordinately regu- lated with BRCA-1 during proliferation and differentiation in mammary epithelial cells. Proc Natl Acad Sci U S A. 93(23):13078-83. Rajan, JV., Marquis, ST., Gardner, HP., Chodosh, LA. (1997). Developmental expres- sion of Brca2 colocalizes with BRCA-1 and is associated with proliferation and dif- ferentiation in multiple tissues. Dev Biol. 184(2):385-401. Rakic, P. (2003) Elusive radial glial cells: Historical and evolutionary perspective. Glia. 43(1):19-32 Rakic, P., Stensas, LJ., Sayre, E., Sidman, RL. (1974). Computer-aided three- dimensional reconstruction and quantitative analysis of cells from serial electron mi- croscopic montages of foetal monkey brain. Nature. 250(461):31-4. Ratineau, C., Petry, MW., Mutoh, H., Leiter, AB. (2002). Cyclin D1 represses the basic helix-loop-helix transcription factor, BETA2/NeuroD. J Biol Chem. 277(11):8847- 53. Razandi, M., Pedram, A., Rosen, EM., Levin, ER. (2004). BRCA-1 inhibits membrane estrogen and growth factor receptor signaling to cell proliferation in breast cancer. Mol Cell Biol. 24(13):5900-13. Rietze, RL., Valcanis, H., Brooker, GF., Thomas, T., Voss, AK., Bartlett, PF. (2001). Purification of a pluripotent neural stem cell from the adult mouse brain. Nature. 412(6848):736-9. Roselli, CE. (1998). The effect of anabolic-androgenic steroids on aromatase activity and androgen receptor binding in the rat preoptic area. Brain Res. 792(2):271-6. Roselli-Rehfuss, L., Mountjoy, KG., Robbins, LS., Mortrud, MT., Low, MJ., Tatro, JB., Entwistle, ML., Simerly, RB., Cone, RD. (1993). Identification of a receptor for gamma melanotropin and other proopiomelanocortin peptides in the hypothalamus and limbic system. Proc Natl Acad Sci U S A 90(19):8856-60.

58 Ruffner, H., Verma, IM.(1997). BRCA-1 is a cell cycle-regulated nuclear phosphopro- tein. Proc Natl Acad Sci U S A. 94(14):7138-43. Russo, AA., Jeffrey, PD., Patten, AK., Massagué, J., Pavletich, NP. (1996). Crystal structure of the p27Kip1 cyclin-dependent kinase inhibitor bound to the cyclin A- CDK2 complex. Nature 382:325-331. Sakakibara, S., Nakamura, Y., Yoshida, T., Shibata, S., Koike, M., Takano, H., Ueda, S., Uchiyama, Y., Noda, T., Okano, H. (2002). RNA-binding protein Musashi family: roles for CNS stem cells and a subpopulation of ependymal cells revealed by tar- geted disruption and antisense ablation. Proc Natl Acad Sci U S A. 99(23):15194-9. Sampei, K., Goto, S., Alkayed, NJ., Crain, BJ., Korach, KS., Traystman, RJ., Demas, GE., Nelson, RJ., Hurn, PD. (2000). Stroke in estrogen receptor-alpha-deficient mice. Stroke. 31(3):738-43. Santagati, S., Melcangi, RC., Celotti, F., Martini, L., Maggi, A. (1994). Estrogen recep- tor is expressed in different types of glial cells in culture. J Neurochem. 63(6):2058- 64. Sasai, Y. (2001). Roles of Sox factors in neural determination: conserved signaling in evolution? Int J Dev Biol. 45(1):321-6. Sauvageot, CM., Stiles, CD. (2002). Molecular mechanisms controlling cortical glio- genesis. Curr Opin Neurobiol. 12(3):244-9. Sawada, H., Ibi, M., Kihara, T., Urushitani, M., Honda, K., Nakanishi, M., Akaike, A., Shimohama, S. (2000). Mechanisms of antiapoptotic effects of estrogens in nigral dopaminergic neurons. FASEB J. 14(9):1202-14. Scheer, N., Groth, A., Hans, S., Campos-Ortega, JA. (2001). An instructive function for Notch in promoting gliogenesis in the zebrafish. Development. 128:1099-1107. Scully, R., Chen, J., Plug, A., Xiao, Y., Weaver, D., Feunteun, J., Ashley, T,. Livingston, DM. (1997a) Association of BRCA-1 with Rad51 in mitotic and meiotic cells. Cell. 88(2):265-75. Scully, R., Anderson, SF., Chao, DM., Wei, W., Ye, L., Young, RA., Livingston, DM., Parvin, JD. (1997b). BRCA-1 is a component of the RNA polymerase II holoen- zyme. Proc Natl Acad Sci U S A. 94(11):5605-10. Scully, R., Chen, J., Ochs, RL., Keegan, K., Hoekstra, M., Feunteun, J., Livingston, DM. (1997c). Dynamic changes of BRCA-1 subnuclear location and phosphorylation state are initiated by DNA damage. Cell. 90(3):425-35. Seri, B., Garcia-Verdugo, JM., McEwen, BS., Alvarez-Buylla, A. (2001). Astrocytes give rise to new neurons in the adult mammalian hippocampus. J Neurosci. 21(18):7153-60. Serrano, M., G.J. Hannon, Beach, D. (1993). A new regulatory motif in cell cycle control causing specific inhibition of cyclin D/CDK4. Nature 366:704-707. Shughrue, PJ., Lane, MV., Merchenthaler, I. (1997). Comparative distribution of estro- gen receptor-alpha and -beta mRNA in the rat central nervous system. J Comp Neu- rol. 388(4):507-25. Shumaker, SA., Legault, C., Rapp, SR., Thal, L., Wallace, RB., Ockene, JK., Hendrix, SL., Jones, BN. 3rd, Assaf, AR., Jackson, RD., Kotchen, JM., Wassertheil-Smoller, S., Wactawski-Wende, J. (2003). Estrogen plus progestin and the incidence of de- mentia and mild cognitive impairment in postmenopausal women: the Women's Health Initiative Memory Study: a randomized controlled trial. JAMA. 289(20):2651-62. Simoncini, T., Hafezi-Moghadam, A., Brazil, DP., Ley, K., Chin, WW., Liao, JK. (2000). Interaction of oestrogen receptor with the regulatory subunit of phosphatidy- linositol-3-OH kinase. Nature. 407(6803):538-41. Smith, AI., Funder, JW. (1988). Proopiomelanocortin processing in the pituitary, central nervous system, and peripheral tissues. Endocr Rev. 9(1): 159-79.

59 Sommer, I., Schachner, M. (1981) Monoclonal antibodies (O1 to O4) to oligodendrocyte cell surfaces: an immunocytological study in the central nervous system. Dev Biol. 83(2):311-27. Song, H., Stevens, CF., Gage, FH. (2002). Astroglia induce neurogenesis from adult neural stem cells. Nature. 417(6884):39-44. Staugaitis, SM., Bernier, L., Smith, PR., Colman, DR. (1990). Expression of the oli- godendrocyte marker 2'3'-cyclic nucleotide 3'-phosphodiesterase in non-glial cells. J Neurosci Res. 25(4):556-60. Takahashi, T., Nowakowski, RS., Caviness, VS. Jr. (1994). Mode of cell proliferation in the developing mouse neocortex. Proc Natl Acad Sci U S A. 91(1):375-9. Temple, S. (2001) The development of neural stem cells. Nature. 414(6859):112-7. Terada, N., Hamazaki, T., Oka, M., Hoki, M., Mastalerz, DM., Nakano, Y., Meyer, EM., Morel, L., Petersen, BE., Scott, EW. (2002). Bone marrow cells adopt the phenotype of other cells by spontaneous cell fusion. Nature. 416(6880):542-5. Thody, AJ. (1999). alpha-MSH and the regulation of melanocyte function. Ann N Y Acad Sci. 885:217-29. Toma, JG., Akhavan, M., Fernandes, KJ., Barnabe-Heider, F., Sadikot, A., Kaplan, DR., Miller, FD. (2001). Isolation of multipotent adult stem cells from the dermis of mammalian skin. Nat Cell Biol. 3(9):778-84. Toran-Allerand, CD. (1991). Organotypic culture of the developing cerebral cortex and hypothalamus: relevance to sexual differentiation. Psychoneuroendocrinology. 16(1- 3):7-24. Toran-Allerand, CD., Singh, M., Setalo, G Jr. (1999). Novel mechanisms of estrogen action in the brain: new players in an old story. Front Neuroendocrinol. 20(2):97- 121. Toyoshima, H., Hunter, T. (1994). p27, a novel inhibitor of G1 cyclin/CDK protein kinase activity, is related to p21. Cell 78:67-74. Tropepe, V., Sibilia, M., Ciruna, BG., Rossant, J., Wagner, EF., van der Kooy, D. (1999). Distinct neural stem cells proliferate in response to EGF and FGF in the de- veloping mouse telencephalon. Dev Biol. 208(1):166-88. Vaccarino, FM., Schwartz, ML., Raballo, R., Nilsen, J., Rhee, J., Zhou, M., Doetschman, T., Coffin, JD., Wyland, JJ., Hung, YT. (1999). Changes in cerebral cortex size are governed by fibroblast growth factor during embryogenesis. Nat Neurosci. 2(3):246- 53. Van der Neut, R., Bar, PR., Sodaar, P., Gispen, WH. (1988). Trophic influences of al- pha-MSH and ACTH4-10 on neuronal outgrowth in vitro. Peptides. 9(5):1015-20. van der Neut, R., Hol, EM., Gispen, WH., Bar, PR. (1992). Stimulation by melanocortins of neurite outgrowth from spinal and sensory neurons in vitro. Peptides. 13(6):1109- 15. Verdi, JM, Schmandt, R., Bashirullah, A., Jacob, S., Salvino, R., Craig, CG., Program, AE., Lipshitz, HD., McGlade, CJ. (1996). Mammalian Numb is an evolutionarily conserved signaling adapter protein that specifies cell fate. Curr. Biol. 6:1134–1145. Verdi, JM., Bashirullah, A., Goldhawk, DE., Kubu, CJ., Jamali, M., Meakin, SO., Lip- shitz, HD. (1999). Distinct human NUMB isoforms regulate differentiation vs. pro- liferation in the neuronal lineage.Proc Natl Acad Sci U S A. 96(18):10472-6. Wakamatsu, Y., Maynard, TM., Jones SU., Weston, JA. (1999). Numb localizes in the basal cortex of mitotic avian neuroepithelial cells and modulates neuronal differen- tiation by binding to Notch-1. Neuron 23: 71–81. Wakamatsu, Y., Maynard, TM., Weston, J.A. (2000). Fate determination of neural crest cells by Notch-mediated lateral inhibition and asymmetrical cell division during gangliogenesis. Development 127:2811–2821.

60 Wang, H., Shao, N., Ding, QM., Cui, J., Reddy, ES., Rao, VN. (1997). BRCA-1 proteins are transported to the nucleus in the absence of serum and splice variants BRCA-1a, BRCA-1b are tyrosine phosphoproteins that associate with E2F, cyclins and cyclin dependent kinases. Oncogene. 15(2):143-57. Wang, L., Andersson, S., Warner, M., Gustafsson, JA. (2001). Morphological abnormali- ties in the brains of estrogen receptor beta knockout mice. Proc Natl Acad Sci U S A. 98(5):2792-6. Wang, L., Andersson, S., Warner, M., Gustafsson, JA. (2003). Estrogen receptor (ER) beta knockout mice reveal a role for ERbeta in migration of cortical neurons in the developing brain. Proc Natl Acad Sci U S A. 100(2):703-8. Wang, Q., Zhang, H., Kajino, K., Greene, MI. (1998). BRCA-1 binds c-Myc and inhibits its transcriptional and transforming activity in cells. Oncogene. 17(15):1939-48. Warbrick, E., Lane, PD., Glover, DM., Cox, LS. (1995). A small peptide inhibitor of DNA replication defines the site of interaction between the cyclin-dependent kinase inhibitor p21WAF1 and proliferating cell nuclear antigen. Curr. Biol. 5: 275-282. Weinmaster, G. (2000). Notch signal transduction: a real rip and more. Curr Opin Genet Dev 10:363–369. Weis, K. (2002). Nucleocytoplasmic transport: cargo trafficking across the border. Curr Opin Cell Biol. 14(3):328-35. Welcsh, PL., Owens, KN., King, MC. (2000). Insights into the functions of BRCA-1 and BRCA2. Trends Genet. 16(2):69-74. Wikberg, JE. Melanocortin receptors: perspectives for novel drugs. Eur J Pharmacol (1999) 375(1-3):295-310. Williams BP, Park JK, Alberta JA, Muhlebach SG, Hwang GY, Roberts TM, Stiles CD. (1997). A PDGF-regulated immediate early gene response initiates neuronal differ- entiation in ventricular zone progenitor cells. Neuron. 18(4):553-62 Wong, KY., Rajora, N., Boccoli, G., Catania, A., Lipton, JM. (1997). A potential mechanism of local anti-inflammatory action of alpha-melanocyte stimulating hor- mone within the brain: modulation of tumor necrosis factor-alpha production by human astrocytic cells. Neuroimmunomodulation 4(1):37-41. Woolley, CS., Gould, E., Frankfurt, M., McEwen, BS. (1990). Naturally occurring fluc- tuation in dendritic spine density on adult hippocampal pyramidal neurons. J Neuro- sci. 10(12):4035-9. Woolley, CS., McEwen, BS. (1992). Estradiol mediates fluctuation in hippocampal synapse density during the estrous cycle in the adult rat. J Neurosci. 12(7):2549-54. Erratum in: J Neurosci 12(10). Wu, LC., Wang, ZW., Tsan, JT., Spillman, MA., Phung, A., Xu, XL., Yang, MC., Hwang, LY., Bowcock, AM., Baer, R. (1996). Identification of a RING protein that can interact in vivo with the BRCA-1 gene product. Nat Genet. 14(4):430-40. Wurmser, AE., Nakashima, K., Summers, RG., Toni, N., D'Amour, KA., Lie, DC., Gage, FH. (2004) Cell fusion-independent differentiation of neural stem cells to the endothelial lineage. Nature. 430(6997):350-6. Xia, Y., Wikberg, JE.(1997). Postnatal expression of melanocortin-3 receptor in rat diencephalon and mesencephalon. Neuropharmacology 36(2):217-24. Xiong, Y., Hannon, GJ. Zhang, H., Casso, D. Kobayashi, R., Beach, D. (1993). p21 is a universal inhibitor of cyclin kinases. Nature 366: 701-704. Yagita, Y., Kitagawa, K., Sasaki, T., Miyata, T., Okano, H., Hori, M., Matsumoto, M. (2002.) Differential expression of Musashi1 and nestin in the adult rat hippocampus after ischemia J Neurosci Res. 69(6):750-6. Yarden, RI., Brody, LC.. (1999). BRCA-1 interacts with components of the histone deacetylase complex. Proc Natl Acad Sci U S A. 96(9):4983-8.

61 Yeh, S., Tsai, MY., Xu, Q., Mu, XM., Lardy, H., Huang, KE., Lin, H., Yeh, SD., Altu- waijri, S., Zhou, X., Xing, L., Boyce, BF., Hung, MC., Zhang, S., Gan, L., Chang, C., Hung, MC.(2002). Generation and characterization of androgen receptor knock- out (ARKO) mice: an in vivo model for the study of androgen functions in selective tissues. Proc Natl Acad Sci U S A. 99(21):13498-503. Ying, QL., Nichols, J., Evans, EP., Smith, AG. (2002). Changing potency by spontane- ous fusion. Nature. 416(6880):545-8. Zhang, H., Somasundaram, K., Peng, Y., Tian, H., Zhang, H., Bi, D., Weber, BL., El- Deiry, WS. (1998). BRCA-1 physically associates with p53 and stimulates its tran- scriptional activity.Oncogene. 16(13):1713-21. Zheng, L., Annab, LA., Afshari, CA., Lee, WH., Boyer, TG. (2001). BRCA-1 mediates ligand-independent transcriptional repression of the estrogen receptor. Proc Natl Acad Sci U S A. 98(17):9587-92. Zhong, Q., Chen, CF., Li, S., Chen, Y., Wang, CC., Xiao, J., Chen, PL., Sharp, ZD., Lee, WH.(1999). Association of BRCA-1 with the hRad50-hMre11-p95 complex and the DNA damage response. Science. 285(5428):747-50. Zhong, W., Feder, JN., Jiang, MM, Jan, LY., Jan, YN. (1996). Asymmetric localization of a mammalian numb homolog during mouse cortical neurogenesis. Neuron 17:43– 53. Zhou, Q., Wang, S., Anderson, DJ. (2000). Identification of a novel family of oligoden- drocyte lineage-specific basic helix-loop-helix transcription factors. Neuron. 25(2):331-43. Zhou, Q., Choi, G., Anderson, DJ. (2001). The bHLH transcription factor Olig2 pro- motes oligodendrocyte differentiation in collaboration with Nkx2.2. Neuron 31(5):791-807. Zimmer, DB., Cornwall, EH., Landar, A., Song, W. (1995). The S100 protein family: history, function, and expression. Brain Res Bull. 37(4):417-29.

62

Acta Universitatis Upsaliensis Comprehensive Summaries of Uppsala Dissertations from the Faculty of Medicine Editor: The Dean of the Faculty of Medicine

A doctoral dissertation from the Faculty of Medicine, Uppsala University, is usually a summary of a number of papers. A few copies of the complete dissertation are kept at major Swedish research libraries, while the sum- mary alone is distributed internationally through the series Comprehen- sive Summaries of Uppsala Dissertations from the Faculty of Medicine. (Prior to October, 1985, the series was published under the title “Abstracts of Uppsala Dissertations from the Faculty of Medicine”.)

Distribution: Uppsala University Library Box 510, SE-751 20 Uppsala, Sweden www.uu.se, [email protected]

ISSN 0282-7476 ISBN 91-554-6111-5