<<

Digital Comprehensive Summaries of Uppsala Dissertations from the Faculty of Pharmacy 69

Neurosteroids Induce Allosteric Effects on the NMDA

Nanomolar Concentrations of Exert Non-Genomic Effects on the NMDA Receptor Complex

TOBIAS JOHANSSON

ACTA UNIVERSITATIS UPSALIENSIS ISSN 1651-6192 UPPSALA ISBN 978-91-554-7114-9 2008 urn:nbn:se:uu:diva-8503                        !  "#$% &  '  &    & ('  ' ) & ('*+ ,'   -     .- '+

 / '   ,+  !+ 0    1    2&& ' 0 3  + 0       & 0    2 0 45  2&& ' 0 3     +    +                     6"+ 76 + + 1.0 "7!4"$4%% 47$$ 4"+

,'          & -& '    '8     '     + ,'      &        '             + ,' &&  &       -'        '  ' 04'44  )0*    ' 4 4      '  $   + ,' '  &     &       + ,'  & ' ' -    &  '8 '   '     ' && &        &      ' 0   + ,'  ' - ' '         & )(.*      & 9:%;.* &&    ' 0    '   ' <  &  ' 0       &   ' '  =      ' 03  + ,' && '            - &'  &       &   +   &       ' (.  9:%;. && 4   -'  05$ !4$% +  &           ).* ' -& &&    + .                 ' ' ' .    -' '  &      '  +  - '     & ' .          '   && & (.  9:%;.  ' 0     '  ' &&  &  (. 9:%;.  '       &  '  $   + ,'   ' - ' '       94&   &  '  $      -'   '  '     &  94&    '  $   + ,'  &&  &  '      '  $          '        -' . + ,' 0                ' 0       &        + ,' &   '       -' &    ' 0             &&      +

   ('       0    03   &                      4 

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

> ,  / '   !

1..0 $6%$46$" 1.0 "7!4"$4%% 47$$ 4"  #  ### 4!% 9 )' #?? +<+? @ A #  ### 4!% 9*

En ny härlig arbetsdag!

List of papers

This doctoral thesis is based on the following original papers, in the text referred to by their Roman numerals (I-V):

Johansson, T, Frändberg, P-A, Nyberg, F and Le Grevès, P. Low con- centrations of neuroactive alter kinetics of 3H- binding to the NMDA receptor in rat the frontal cortex. Br J Pharmacol 2005 (146) 894-902 Johansson, T, Nyberg, F and Le Grevès, P. Allosteric modulation by neurosteroids and the impact of temperature on 3H-ifenprodil binding to NR1/NR2B receptors. Manuscript submitted to Br J Pharmacol 2008 Johansson, T, Frändberg, P-A, Nyberg, F and Le Grevès, P. Molecular mechanisms for nanomolar concentrations of neurosteroids at NR1/NR2B receptors. JPET 2008 (324) 759-768 Johansson, T, Elfverson, M, Birgner, C, Frändberg, P-A, Nyberg, F and Le Grevès, P. Neurosteroids alter glutamate-induced changes in neurite morphology of NG108-15 cells. J Biochem Mol Biol 2007 (104) 215-219. Johansson, T, Zhou, Q, Nyberg, F and Le Grevès, P. Alteration in the action at the sigma 1 receptor but not at the NMDA receptor after chronic AAS administration. In manu- script.

All papers are written by the first author in association with co-authors. In paper I-III & V the first author are responsible for all analyses and the major part of the laboratory work. In paper IV, analyses were shared between the three first authors and laboratory work was performed by two first authors. The planning of the experiments was a co-operation between all authors (paper I-III), first, second and last author (paper IV) and first, third and last author (paper V). Reprinted with permission of Nature Publishing Group (paper I), the AS- PET journals (paper III) and Elsevier Ltd (paper IV).

Contents

Introduction...... 11 The definition of neurosteroids and their biosynthesis...... 11 Functional significance of neurosteroids...... 14 and ...... 14 Mood...... 14 Neurodegeneration and ...... 15 The rapid effects of neurosteroids in the central ...... 17 Anabolic androgenic steroids ...... 17 The NMDA receptor ...... 18 Composition, and distribution ...... 18 Central processes involving the NMDA receptor...... 20 The of the NMDA receptor...... 20 The ...... 21 Classification and distribution ...... 21 Central processes involving the sigma receptor ...... 22 The pharmacology of the sigma receptor ...... 22 Ligands as tools for the study of NMDA and sigma receptors ...... 23 Ifenprodil ...... 23 ...... 24 Blocking agents ...... 24 Aims of this thesis...... 26 Specific aims ...... 26 Methods ...... 27 General procedures...... 27 Animals and treatment ...... 27 Dissection ...... 27 Cells...... 28 culturing ...... 28 CHO-E2 cells...... 28 NG108-15 cells...... 29 Membrane preparation ...... 30 Rat tissue membrane preparation for receptor binding (paper I & V).30 Cell membrane preparation for receptor binding (paper II & III) ...... 30 Ligand binding ...... 30

Ligand binding calculations ...... 31 Saturation studies...... 31 Competition studies ...... 32 Dissociation studies ...... 33 Fluorescence measurements (paper III) ...... 34 2+ Cell preparation for [Ca ]i measurements ...... 34 Loading of fluorescence ...... 34 2+ [Ca ]i measurement ...... 34 Neurite measurement (paper IV)...... 35 Data analyses...... 35 Results and discussion ...... 36 Optimizing the processes ...... 36 Evaluation of the membrane preparation...... 36 The discrimination of receptors (paper I-III & V)...... 36 The effects of neurosteroids involving the NMDA receptor...... 37 Allosteric modulation of the 3H-ifenprodil binding by neurosteroids (paper I-III & V) ...... 37 Different targets for PS and 35S (paper I-III) ...... 38 The neurosteroid effect curves are bell- or U-shaped (paper I-III)...... 40 Temperature involvement (paper II)...... 41 Allosteric & direct effects of neurosteroids (paper I-III & V)...... 42 The effects of neurosteroids involving the sigma receptor (paper IV-V).45 Influences of AAS on neurosteroid effects (paper V)...... 45 Imagining the use for neurosteroids ...... 47 Neurosteroids in combating drug abuse...... 47 Antidepressive and use of neurosteroids ...... 47 Neuroprotective use of neurosteroids ...... 48 Conclusions...... 50 Future perspective...... 52 Populärvetenskaplig sammanfattning ...... 54 Acknowledgements...... 56 References...... 58

Abbreviations

S sulfate 3-HSD 3-hydroxysteroid dehydrogenase-isomerase 3H-ifenprodil tritium labeled ifenprodil AAS anabolic androgenic steroids ALLOPREG ,  PROG ALLOPREGS allopregnanolone sulfate, S P  -amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid ANOVA analysis of variance APV DL-2-amino-5-phosphonopentaoic acid Bmax Maximum ligand binding CHO chinese hamster ovary CNS central nervous system D-MEM Dulbecco’s modified Eagle’s medium DHEA DHEAS dehydroepiandrosterone sulfate DMSO dimethylsulphoxide DTT dithiothreitol EPSC excitatory post-synaptic current FBS foetal bovine serum Fura-2 AM fura-2 acetomethoxy ester GABAA gamma-amino butyric acid A GBR 12,909 1-[2-[bis(4-fluorophenyl)methoxy]ethyl]-4-[3- phenylpropyl]piperazine GBR 12,935 1-[2-(diphenylmethoxy)ethyl]-4-(3-phenylpropyl)- piperazine HAT hypoxanthine, aminopterin, thymidine IC50 inhibitory concentration 50% icv intracerebroventricular Kd apparent dissociation constant Ki inhibition constant Koff dissociation rate constant Kon association rate constant MK-801 (±)-5-methyl-10,11-dihydroxy-5h-dibenzo (a,d)cyclohepten-5,10-imine) a.k.a. MPTP 1-methyl 4-phenyl 1,2,3,6-tetrahydropyridine NMDA N-methyl-D-asparatate

nH the Hill coefficient NPC Niemann-Pick C type NR1 NMDA receptor subunit 1 NR2 NMDA receptor subunit 2 NR2A NMDA receptor subunit 2A NR2B NMDA receptor subunit 2B NR3 NMDA receptor subunit 3 PREG PROG PS PTX pertussis toxin (+)-SKF 10,047 (+)-N-Allylnormetazocine SMD steroid modulatory binding site SNRI and noradrenalin re-uptake inhibitor SSRI selective serotonin re-uptake inhibitor VTA Ventral tegmental area

Introduction

Steroid hormones are powerful mediators in the body. The findings that ster- oids not only act via nuclear receptors affecting the gene transcriptions, but also directly at membrane bound receptors, could both explain the rapid ef- fects of steroids reported and present possible targets for treatment of dis- eases and pathological conditions.

The definition of neurosteroids and their biosynthesis Neurosteroids are defined as steroid compounds, synthesized in and inducing rapid effects on the central nervous system (CNS). Neuro-active steroids are referring to all steroids with effects on the CNS. Neurosteroids are derived from or as metabolites from other steroids. They exert both rapid and slow effects. Rapid effects are thought to be mediated via membrane bound receptors, a mechanism apart from the classical, slower genomic mechanism 16. In the CNS, six dominating neurosteroids have been identified: PROG and its metabolites allopregnanolone (ALLOPREG or ) and preg- nanolone sulfate (35S) (Figure 1) are considered as inhibitory neuroster- oids. Pregnenolone (PREG), Dehydroepiandrosterone (DHEA) (Figure 2) and their respective sulfate (PS (Figure 2) and DHEAS) are stimulatory. PS and DHEAS are more active than the non-sulfated parent steroids 205.

Figure 1. The structures of three inhibitory neurosteroids, PROG (left), ALLOPREG (middle) and 35S (right).

11

Figure 2. The structures of two stimulatory neurosteroids, PS (left) and DHEA (right).

Enzymes that form PREG from cholesterol and other neurosteroids from PREG include two cytochrome P450s, 3-hydroxysteroid dehydrogenase- isomerase (3HSD), 3-hydroxyoxidoreductase and 5-reductase 16,53,197,199. Sulfotransferase and sulphatase add respectively remove the sulfate in 3- position 199 (Figure 3). These enzymes are expressed in brain regions such as the hippocampus, the cortex and the 53,197.

Figure 3. (See next page) The synthesis of neurosteroids and the reversible metabo- lism. * indicates a pathway shown in vitro but still not in vivo.

Neurosteroids, and sulfated neurosteroids in particular, have been re- ported to be present at low amounts in the rat brain 117,165. Nevertheless, fem- tomoles of PS administered by intracerebroventricular (icv) injections have in different studies on rodents been demonstrated to affect processes involv- ing the NMDA receptor 79,80,180,181,201,314. Examinations of whole brain have found around 6 ng/g PS in rat 335 and in mice 309, but also 20 times lower amounts has been reported 117,165,269. In vitro assays have demonstrated that certain neurosteroids exert anti-apoptotic actions in concentrations around 1 nM 43. The effects of DHEA and DHEAS persist in pM concentration on neuritogenesis 197.

12

13 Functional significance of neurosteroids

Memory and learning Neurosteroids are involved in cognitive performance and memory consolida- tion 16. Neurosteroids administrated exogenously show anti-amnesic and promnesic properties on memory and learning. The ability of learning and remembering things and tasks and their decline over the age is correlated to endogenous levels of neurosteroids and their decline 188,205. Memory and cognitive performances in rodents can be enhanced by PS 79,80,201 and DHEAS 81,82. PS is also shown to reverse NMDA antagonist-induced mem- ory deficits 180,181.

Mood Neurosteroids are shown to be involved in mood and temper. The female sexual behaviour in rats is believed to be initiated by PROG, or more potently, by the neurosteroid ALLOPREG. It is regulated by neurosteroid interactions with NMDA receptors in the ventral tegmental area (VTA) and in the hypothalamus and can be banished by the NMDA receptor antagonist MK-801 237. Lowered levels of ALLOPREG are correlated to 241 and women diagnosed with premenstrual dysphoric disorder have reduced serum levels of ALLOPREG compared to controls 93. PROG is also targeting the sigma receptor, acting as an antagonist while its metabolites ALLOPREG and pregnanolone are acting as positive modula- tors of the GABA receptor 205. ALLOPREG is described to possess anxio- lytic properties and its GABAA stimulation effects are believed to be respon- sible for this 30. Social isolation decreases the 5-reductase, an enzyme cru- cial in facilitating the conversion of PROG to ALLOPREG, in the hippo- campus and in glutaminergic in the frontal cortex, and the amygdala, resulting in symptoms in mice as aggression and 3. ALLOPREG levels are lowered as a consequence of intake 223 and as well during as and panic disorders. The levels can be restored by selective serotonin re-uptake inhibitors (SSRI) as 243,260,310 and 310. Fluoxetine has also been recorded to re- establish levels of the neuroactive steroids 35 and 35 260. Norfluoxet- ine, another SSRI, has been shown to re-establish lowered ALLOPREG lev- els in doses too low for efficiently inhibit the serotonin reuptake 242, an effect that is proposed to be mediated by sigma 1 receptors 263. Depressive behaviour in aged mice is believed to be the result of the de- cline of neurosteroids. It can be reversed by , a selective sigma 1

14 receptor , in a dose-response manner 238. Treatment with PS or DHEAS reversed the depressive symptoms to levels of control, in a depres- sion model, using adrenalectomized and castrated male mice 189.

Neurodegeneration and neuroprotection Neurodegeneration occurs as a result of apoptosis or necrosis. It is induced by neurochemical modulators that cause injuries to the nervous system. One common injury is the NMDA receptor mediated 311. Neurode- generation is characterized by neuronal cell death and a progressive dysfunc- tion. It is a core problem in diseases as Alzheimer’s disease, non- Alzheimer’s dementia, Parkinson’s disease, and 332. Neuro- protection is defined as the effect that prevents or recovers neural systems from neurodegeneration 311. Key proteins involved in Alzheimer’s disease, such as tau and -amyloid, regulate the synthesis of neurosteroids in vitro synthesis. 266,311. A mutation in the gene for tau changes the involvement of the modified tau in neuroster- oid synthesis. The data suggests that the neurodegeneration seen in Alz- heimer’s disease are due to lowered production of certain neurosteroids 266. PREG has in vitro been shown to protect cells from -amyloid toxicity 103 and in vivo to compensate for the modification (hyperphosphorylation) of tau 17. In patients diagnosed with Alzheimer’s disease, PS and DHEAS were shown to be significantly lowered compared to non-dement controls, in cer- tain brain regions 317 and ALLOPREG was lowered in the frontal cortex 178 as well as in plasma 23,279. These findings were supported by a negative cor- relation between cortical -amyloid and the lowered PS levels 317. A re- corded consequence of lowered levels of neurosteroids in patients suffering from Alzheimer’s disease is aggression 240, a symptom seen in other diagno- ses as well, see sections below. The NMDA receptor could very well be involved in these mechanisms, since site are shown to counteract memory impairment in- duced by -amyloid 187 and numerous reports are available considering the diseases influence on the NMDA receptor 39,89,133,230,282,307,308. Parkinson’s disease is characterized by the loss of dopaminergic neurons in substantia nigra. This state is mimicked in in vivo experiments by the ad- ministration of MPTP, a substance that bring damage to those neurons selec- tively. DHEA as well as 17- have the ability to prevent the devel- opment of MPTP induced Parkinson’s disease, in a neuroprotective manner 38,62,97. Also PROG has shown neuroprotective effects against MPTP assault on neurons in the striatal region 38. PROG and its metabolite 19-norprogesterone can protect primary hippo- campal cultures against glutamate induced toxicity 218. ALLOPREG

15 show neuroprotective effects in a Niemann-Pick C type disease (NPC) model 99. The NPC is an inherited neurodegenerative disease with parallels to Alzheimer’s disease in the cellular pathology 220. Hypoxia is a certain way to induce neuronal damage. It can be found in the brain as well as in the spinal cord, e.g. after stroke or physical injury. Neurosteroids as DHEAS, the DHEAS metabolites 7-OH- or 7-OH-epiandrosterone 134,209 are all shown to exert neuroprotection in hypoxia 138,154,247. PS and 35S are suggested to be useful in stroke therapy 281. In the fetal brain the levels of ALLOPREG is much higher than in the newborn brain. It is believed that ALLOPREG induces a sleep-like state in the fetus. But ALLOPREG and other neurosteroids also protects the fetal brain from injury, from e.g. hypoxia 120. The fetus also responds to hypoxia with an up-regulation of ALLOPREG 26. PS is crucial in the neuronal plas- ticity in newborns, acting via -amino-3-hydroxy-5-methyl-4- isoxazolepropionic acid (AMPA), receptors and NMDA receptors. NMDA receptors in newborns are constituent of NR1/NR2D (see the part below about the NMDA receptor) presynaptically and NR1/NR2B postsynaptically 175. Cortical neurons from fetus, growth in vitro, change morphology as a direct response to DHEA or DHEAS; DHEA induce the axons to grow and DHEAS the dendrites to grow 197. On the other hand, age tends to display a decline in neurosteroid levels, in particular PROG 238, the precursor to ALLOPREG. DHEA and its sulfate ester DHEAS are involved in the processes of aging 102,161 where levels of DHEA and DHEAS, both with antiglucocortical effects, decrease and levels of remain unaltered compared to young adults 2,102,160. The declina- tion of DHEA and DHEAS levels results in levels for a 70 years old man at only about 25% the levels of a 25 year old. The two DHEA derivates 7- OH-DHEA and 7-OH-DHEA both show neuroprotective properties, which are believed to be mediated by their counteraction of 209. This has been speculated to be subject for a diagnose as DHEA and DHEAS deficient and therefore be subject to treatment with supplements 2. DHEA and DHEAS are currently being sold on the US market as an anti-age drugs and clinical experiments have confirmed some positive results for the two neurosteroids as “youth drugs” 15,208. In vitro experiments have shown that neurosteroids as DHEAS, DHEA and ALLOPREG, are important mediators in aging, counteracting apoptosis 43 a process they can not keep inhibiting when levels are decreased with age 44.

16 The rapid effects of neurosteroids in the central nervous system

Neurosteroids interact with and modulate membrane bound receptors, such 19,153,171 191 as the -amino butyric acid A (GABAA) , the sigma 1 and the N- methyl-D-aspartate (NMDA) receptor 123,132,231,281, mechanisms which as- sumably mediate their non-genomic effects. There data presented in this field is still limited and most of the data originates from electrophysiological experiments. Electrophysiological experiments using high concentrations (micromolar) of the neurosteroids, detect modulatory effects on recombinant NMDA receptors. In those studies, pregnenolone sulfate (PS) (Figure 2) acts as a positive , stimulating the agonist induced response of NMDA receptor complexes composed by NR1/NR2A or NR1/NR2B subunits 174,333. 35S is reported to exert inhibitory action in those studies 174. Similar effects of PS and 35S at micromolar concentrations were also found when Ca2+-influx was measured in CHO (Chinese hamster ovary) cells stably expressing the NR1/NR2B subtype of the NMDA receptor 211.

Anabolic androgenic steroids Anabolic androgenic steroids (AAS) are the male and synthetic derivates of testosterone (Figure 4). They were originally de- veloped for therapeutic purposes, but nowadays also commonly used by athletes 164,336 or by criminals 149,292. Still, testosterone and its ester derivates with prolonged duration are clinically used to treat male hypogonadism as replacement therapy 280. The testosterone derivate nandrolone (Figure 4) is being used in terminal diseases to promote well-being and increase appetite as well as to counteract the itching of chronic biliary obstruction. Nandro- lone is also used to assist the recovery after severe burns or postoperative wounds. It is usually administrated as its ester nandrolone decanoate (Figure 4), which creates a slow-release depot in vivo, from where nandrolone is released 13,252.

Figure 4. The structures of the three commonly used AAS’s testosterone (left), nan- drolone (middle) and nandrolone decanoate (right).

17 The illegal use of AAS include the risk of side-effects from suprathera- peutic doses, including influence on the genital system and impotence 25,78, depression 246,293, anxiety 257,258, cardiac failure 67,236,290, even though there could be other reasons for cardiac diseases in this group, such as being target for hostility 250 or anger attacks 75,76 shown to increase the risk for cardiac disease. Neither is the risks of being infected with lethal virus infections, as HIV or hepatitis C, negligible in the illegal use of AAS 4. The desired effects of AAS in supraphysiological doses are increased muscle mass and strength 24, aggression 35,235,239,291 and self-confidence 72. The aggressive behavior following AAS treatment is accompanied by lowered levels of ALLOPREG in the mouse brain 241. This, both ALLOPREG levels and aggression could be reversed by administration of fluoxetine 77,121 or its metabolite norfluox- tetine 77,121,244 or by the administration of and PROG 242. AAS are believed to interfere with several receptor systems within the CNS, e.g. the NMDA receptor. Chronically administered AAS changes re- ceptor expression, while acutely they can act as allosteric modulators (for review see 50). Side effects of AAS are suggested to be mediated by interac- tions between AAS or its metabolites and other steroid hormones 12. Through the rapid, allosteric modulation, AAS or metabolites of AAS, act as neuro- active steroids 83, or interactions between AAS and neurosteroids could exist.

The NMDA receptor

Composition, function and distribution Glutamate is the endogenous ligand of two major groups of receptors, the G- protein coupled metabotropic receptors and the ligand-gated ion channels. The latter ones are named after ligands, which selectively activate the recep- tor. These are the AMPA, the kainate and the NMDA receptors 322. The NMDA receptor is blocked by Mg2+ in a voltage dependent manner. When activated by glutamate and the co-agonist glycine the blockade is removed and the ion-channel is permeable for Ca2+, Na+ and K+ 192. The NMDA receptor is composed out of two pairs of subunits, two NR1 and two NR2 subunits. The NR1s are either NR1a or NR1b, while the NR2Bs are a homologue or heterologue combination of the four available (NR2A-D) subunits 206,207 (Figure 5). Each subunit consists of four trans- membrane units connecting the extracellular N-terminal with the intracellu- lar C-terminal (Figure 6). The combination of subunits determine the spe- cific pharmacological and functional properties of the receptor 322.

18

Figure 5. The NMDA receptor is constructed out of two pairs of subunits, two NR1s and two NR2s, forming the ion pore. The four proteins forming the receptor are situated in the cell membrane.

Figure 6. Each one of the four subunits forming the NMDA receptor is anchored in the cell membrane with four transmembrane units, M1-M4, connecting the extracel- lular N-terminal with the intracellular C-terminal. M1-M3 form the ion pore 228.

Different brain regions are hosting differently composed NMDA receptors. The NR1 subunits are distributed throughout the whole CNS, and the NR2 subunits are differently distributed in the CNS, NR2A and NR2B dominating

19 in cortex and the hippocampus 206,207 and NR2B is also present in the hypo- thalamus 217, while NR2A is found in all brain regions 312. The distribution of differently composed NMDA receptors is also changed during development 274. The affinity for certain ligands is also al- tered during development 143. The NR2B subunit is to be found all over the embryonic brain, but in the postnatal brain NR2B subunits is restricted to the forebrain 217,312, the hippocampus and the hypothalamus 217. The NR1 and NR2D subunits are expressed unaltered during the brain development, NR1 all over the brain and NR2D in diencephalon and the brainstem. The NR2A and NR2C subunits both appear postpartum, the NR2A in all brain regions 312 and NR2C in the cerebellum 63,312. The changes during development are believed to be crucial for the synaptic plasticity 175.

Central processes involving the NMDA receptor Uncontrolled or excessive activation of the NMDA receptor leads to neu- rodegeneration, cell death and epilepsy, whereas impaired control of its acti- vation is thought to be involved in schizophrenia and neurodegenerative 48,69,98,337 221,233,299-303,319 diseases and also in drug addiction . It is also in- volved in memory and memory consolidation 169,276. An impairment or down-regulation of the NMDA receptor decreases spatial learning and other memory functions and pharmacological blockade has been shown to be neu- roprotective, , amnesic and anticonvulsative 85,152,226,227. The NMDA receptor antagonist improves mood and cognitive functions in patients suffering from Alzheimers’s disease 9 and memantine 283, ifenprodil and haloperidol 245 have been proposed to display neuroprotection.

The pharmacology of the NMDA receptor The NMDA receptor has several well-defined binding sites for a various number of ligands, endogenous as well as exogenous substances. This in- clude sites for the agonist glutamate 115,131,196,202 and the co-agonist glycine 32,137 and for polyamines 259,325,326, ifenprodil 321, protons 289,297, neurosteroids 124,125,127,128,132,136,231,232, ions 150,249, ions 52,107,192,222, MK-801 71 (for review, see 68) and site for reduction/oxidation 5,150. The glycine site is located exclusively on the NR1 subunit 119,151 and the others on different NR2 subunits or in the . However, it is described as many as three different sites for polyamines, and one of those is proposed to be situ- ated on the NR1 subunit and increase the glycine binding. The other two are described to be located on the NR2B subunit, one of them is located near the ifenprodil site, which decreases the glutamate binding. The other one is reck- oned as a glycine independent stimulation 323. The polyamine site on the NR1 subunit has been suggested to be accompanied by a separate ifenprodil binding site as well, both regulating the glycine binding 253.

20 It is also known that several of those ligands interact with each other when binding to the receptor: the main agonist glutamate increases the bind- ing of ifenprodil and vice verse. Ifenprodil and polyamines decrease each other’s interaction with the receptor, to such an extent it once was believed they shared the same site 41,142,256,268. Ifenprodil and polyamines are shown to induce their effects via increasing respectively decreasing the tonic proton inhibition of the receptor 210,298. Side effects from NMDA receptor antagonists, e.g. the MK-801, limit their clinical usefulness. This is quite understandable due to the receptor distribution throughout the central nervous system (CNS) and the non-selectivity of the actual drugs. Subtype selectivity was asked for and has lead to the development of subunit selective antagonists. However, these compounds also proved to carry too many side effects. The all-or-nothing effect of channel opening, as occurs with competitive antagonists or high- affinity channel blockers, seems to be too powerful for clinical use. The ex- istence of several modulatory sites, where allosteric modulators regulate the effect of endogenous agonists, shows promises of being therapeutic com- pounds 322. Such group of substances includes the neurosteroids. An allos- teric ligand enjoys several therapeutic advantages over conventional ligands. These include that allosteric ligands not being active other then when the receptor also bind the endogenous ligand. The allosteric ligand then attenu- ates or potentiates its signal. The effects of the allosteric ligand will follow the synaptic release of endogenous ligands, the release pulses and therefore better mimic a natural state. Treatment with allosteric ligands does not cause desensitisation or internalisation of the receptor, which can be a problem with conventional ligands. Thus will the effects of allosteric ligands persist over the treatment. The binding site for conventional ligands often share similarities with other receptors, as a result of the evolution. The conven- tional ligands will therefore bind to other targets as well, causing side- effects. The allosteric site seems to be much more specific for the allosteric ligand, why side-effects are far less expressed. The allosteric effect is also highly saturable, reducing the risk for overdosing and side-effects 135.

The Sigma receptor

Classification and distribution The sigma receptor is an atypical receptor 271 associated with the NMDA receptor and also an important target for neurosteroids 205. Progesterone is believed to be the endogenous ligand 91,191. It was first defined as an opioid receptor 176, but has then proven to be a completely different type of receptor family 284,287. The sigma receptor family is divided in two subtypes 248, sigma

21 1 and sigma 2, where the sigma 1 receptor is far more studies and better characterized than the sigma 2 receptor. The sigma 1 receptor is classified as 130,295 a Gi receptor, since it is sensitive to pertussis toxin (PTX) , but shown little similarity with other G-protein receptors 91. The sigma receptors are present all over the rat CNS including the cortex 6,34,56. The distribution of the two sigma receptor subtypes are quite equal in the rat brain, apart from the motor cortex, which is only inhabited by sigma 2 receptors, a region responsible for locomotion 34. Also the nucleus accumbens and the substan- tia nigra predominantly host sigma 2 receptors, whereas the density of sigma 1 receptors was higher in the dentate gyrus and some nuclei of thalamus and hypothalamus 34.

Central processes involving the sigma receptor The sigma receptor mediates dysphoric and hallucinogenic effects. The dys- phoric properties can be used in antidepressive treatment 182,191,277,278,309. The sigma receptor is also believed to mediate the change in mood in pregnant women 21. The sigma receptor is also involved in memory function as sigma ligands promote anti-amnesic or pro-mnesic effects 183,185,188,190.

The pharmacology of the sigma receptor The effects of sigma receptors are mediated by controlling the mobilization of intracellular calcium 177. They cause dose dependent increase in intracellu- lar calcium via the sigma 2 receptor, probably by releasing Ca2+ from intra- cellular storages. The raise in intracellular calcium is assumed to be the sec- ond messenger signal to the sigma 2 receptor activation 328. Sigma receptors also control intracellular calcium by modulation of NMDA receptor re- sponses 20,22,177,204. The modulation of the NMDA receptor can be shown as counteracting of effects, e.g. glutamate or NMDA induced postsynaptic re- sponse 51,294 or excitotoxicity 275, as changes in calcium influx 112 or in changes in binding of NMDA receptor ligands, e.g. 3H-MK-801, using selec- tive sigma ligands without affinity to the NMDA receptor, as MS-377 139, (+)SKF 10,047 and (-)PAPP 275. This can be translated into functional stud- ies, where selective sigma ligands are also shown to attenuate MK-801 in- duced learning impairment 184,190.

22 Ligands as tools for the study of NMDA and sigma receptors

Ifenprodil Ifenprodil (Figure 7) is a compound, with affinity to many receptors, among them the NMDA receptor and the sigma receptor. It was originally developed as a vasodilating agent, exploiting its affinity for 40,104,170 57 1-adrenergic receptors and used as a cerebral vasodilator and tested as anti-ischemic agent in models for cerebral ischemia 95. Its affinity for the NMDA receptor was revealed in the 1980s. Ifenprodil acts neither as a channel blocker, nor as an antagonist to glutamate or glycine sites, why it is classified as a non-competitive NMDA receptor antagonist 159,256,321. Ifen- prodil has its binding site on the NMDA receptor solely on the NR2B sub- unit and remains the best characterized of the subunit specific NMDA recep- tor ligands. It was discovered that ifenprodil possesses neuroprotective prop- erties, while it lacks several of the side effects shown by classic NMDA re- ceptor antagonists. Ifenprodil was first addressed as a polyamine antagonist 18,41,268,288, but further studies showed that the interaction between poly- amines and ifenprodil is allosteric. That allosteric effect results in highly decreased affinity for the other ligand 86,87,142. However, due to its affinity to the sigma receptor 54,108, the piperazine acceptor site 59, the voltage- 2+ 14,49 11,194 dependent Ca channels and the 5-HT3-receptor , ifenprodil was doomed to never enter the clinical area, instead forever being a good phar- macological tool.

Figure 7. The structure of the NR2B selective agent ifenprodil.

In the beginning ifenprodil was defined to have a 100-fold selectivity for NR1a/NR2B over NR1a/NR2A 321, but later studies claim that NR1a/NR2A, NR1a/NR2C and NR1a/NR2D all are almost insensitive to ifenprodil 63,100,324. Transgenic animal techniques using knockout mice further show that NMDA receptors on hippocampal neurons lacking the NR2B subunit do not respond to ifenprodil 296. 3H-ifenprodil, a commonly used radioligand for in vitro experiments on NR2B, shows affinity for several other binding sites in rat brain membranes. To ascertain that the NR2B binding really is being studied, the other high affinity targets must be blocked. The high affinity sites include, apart from

23 NR2B, the sigma receptor 109,111,140, which can be blocked by GBR 12,909 55,59,101, the piperazine acceptor site, which is blocked by trifluoroperazine 59. Trifluoroperazine also blocks the sigma receptor 59,331 as well as the last high 304 affinity target, the 1-adrenergic receptors . The pure binding to the NMDA receptor is then dose-dependently biphasic 217. The high affinity interaction is the one between the ifenprodil binding site on the NR2B sub- unit. The explanation for the low affinity interaction could be found in the putative interaction between ifenprodil and the glycine site on the NR1 sub- unit 159,253,321.

Haloperidol Haloperidol (Figure 8) (paper II-III & V) is a widely used anti-psychotic agent 252 with positive effects on depression and social disabilities in chronic schizophrenia 195,261. The anti-psychotic effects are believed to reside from blocking dopaminergic receptor. Apart from the receptor affinity, haloperidol display binding characteristics reminding of that for ifenprodil. Haloperidol binds to NR2B 60,86,122,126, serotonin and -adrenergic receptors 252 and sigma receptors 56,111,287,313. Haloperidol actually once was considered as the prototypic drug for the sigma receptor 130,203,284. It was used to dis- criminate the sigma site from the PCP-binding site on the NMDA receptor 129,130,287. Haloperidol was used in order to assure the 3H-ifenprodil binding to be to the NR2B target (in combination with trifluoroperazine) and in the sigma receptor assay.

Figure 8. The structure of haloperidol, which shares several binding sites with ifenprodil.

Blocking agents The GBR 12,900-family is a series of compounds originally designed to block dopamine transporters, inhibiting the dopamine uptake, but was found to also label other targets. GBR 12,935 (Figure 9a) and GBR 12,909 (Figure 9b) are commonly used to isolate the NMDA receptor site in various binding assays. They share structure similarities with both ifenprodil (Figure 9c) and haloperidol (Figure 9d). GBR 12,935 is used preferably to label the piperazine acceptor site 8,94 and GBR 12,909 labels the sigma receptor 37,55.

24 Their selectivity is however not utterly, why too high concentrations of GBR 12,935 would interfere with the sigma receptor binding, elucidated in the optimization process of the 3H-ifenprodil binding to the sigma receptor. An- other ligand used to isolate the NMDA receptor site is trifluoroperazine 59 (Figure 9e).

Figure 9. The structures of (a) ifenprodil, (b) haloperidol, (c) GBR 12,935, (d) GBR 12,909 and trifluoroperazine show similarities, as do some of their receptor affini- ties.

25 Aims of this thesis

The rapid physiological effects of neurosteroids are to quite an extent exam- ined, but the molecular mechanisms underlying their actions are still not fully elucidated. The aim of this thesis was to increase the understanding of the mechanisms behind neurosteroid mediated modulation of NMDA and sigma receptors, to bring clarity and knowledge to the field.

Specific aims Characterisation of the influence of PS and 35S on ifenprodil binding to rat cortical NMDA receptors (paper I). Characterisation of the influence of PS and 35S on ifenprodil inhibitory action on glutamate stimulated calcium influx and ifenprodil binding re- combinant NMDA receptors (paper II & III). Investigate the impact of temperature on ifenprodil binding and the action of neurosteroids on such binding to the recombinant NMDA receptors (pa- per II) Examine the effects of nanomolar concentrations of PS, 35S and ifen- prodil on glutamate stimulated neurite outgrowth in vitro (paper IV). Investigate the consequences of in vivo AAS (nandrolone decanoate) treat- ment on the effects of nanomolar concentrations of PS and 35S on NMDA and sigma receptors in rat cortical membranes (paper V).

26 Methods

General procedures All experiments were carried out at the division of research on biological drug dependence, Biomedical center, Uppsala, Sweden. Animal experiments were conducted in the animal facilities. In vitro experiments were repeated at least three times on different days.

Animals and treatment Adult male Sprague–Dawley rats (Alab AB, Sollentuna, Sweden) weighing 200–220 grams (approximately 11-week-old) at the start of the studies, were housed in groups of four in air-conditioned rooms under an artificial 12 hours light-dark cycle at a temperature of 22-23°C and a humidity of 50- 60%, food (R36 standard pellets) and water ad libitum. The cages used elm chips as surface. Prior to tissue sampling or treatment, the rats were allowed to adapt to the laboratory environment for one week. The animal experimen- tal procedure was approved by the local experimental animal committee in Uppsala, Sweden. In paper I the animals received no treatment before tissue sampling, in paper V the animals were randomly divided into two groups, one receiving nandrolone decanoate (15 mg/kg) every third day and the other group re- ceived vehicle (arachidonis oleum). The treatment lasted for three weeks. The injections were administered subcutaneously in volumes of 100 μl. The sites of injection were shifted, never injecting the same location twice, in order to avoid local skin reactions.

Dissection The animals were sacrificed by decapitation. Brain tissues were rapidly re- moved from the skulls and placed on ice. The frontal cortex, the cortex, the hippocampus and the hypothalamus were dissected, immediately frozen and stored at -80°C until further processed.

27 Cells

Cell culturing Cells were grown in a controlled environment with a humidified atmosphere containing 5% CO2 at 37 C (Sanyo MCO-15AC CO2 incubator, Sanyo E&E Europe BV, Holland).

CHO-E2 cells The CHO-E2 cell line (Figure 10) (paper II & III) is a cell line genetically enhanced by Uchino and co-workers (2001). It was a kind gift of Dr. Shigeo Uchino 305. It is carrying the genes for functional NMDA receptor [NR1/NR2B]. The CHO cell line is not capable to cope with such massive expression of NMDA receptor, why the genes are under the control of the Drosophila heat-shock protein promoter 70. Activation of the promoter is carried out by heat induction for 2 hours. The receptors are then present after 6 h for another 6 h. After the heat induction, the cells need efficient NMDA receptor blockade to survive until used in assay, provided by APV.

Figure 10. The CHO-E2 cells as seen in the microscope, look like any ordinary CHO cell, but possess the ability to express fully functional NMDA receptors, formed from two NR1s and two NR2Bs.

The CHO-E2 cells were grown on 100 mm culture dishes (Sarstedts, Nürnbrecht-Rommelsdorf, Germany). The culture medium (Dulbecco’s modified Eagle’s medium (D-MEM) supplemented with 10% foetal bovine serum, 1.2 mg/ml geneticin, 2 μg/ml blasticidin S HCl and 10 μg/ml puro- mycin) was changed every sixth day. The cells were grown to approx. 80% confluence and split 1:3, generally every sixth day, using trypsine/EDTA (0.05%/0.02%), before transferring to fresh 100 mm culture. Cells at pas- sages 5 – 30 were used.

Heat induction of CHO-E2 To induce NMDA receptor expression in the CHO-E2 cells, culture dishes containing 150000 cells cm-2 were incubated for 2 h at 43 C in a controlled humidified environment containing 5% CO2 (Micro Galaxy, Zander Medical

28 Supplies, Vero Beach, FL, USA) and were then maintained at 37 C for 6 h, in order to express the proteins, in culture medium supplemented with 1 mM APV, before used in calcium assays or harvested for membrane preparation.

NG108-15 cells The neuroblastoma x glioma NG108-15 hybrid cell line (Figure 13) (paper IV) was employed due to its ability to produce axon-like neurites as a result from elevated intracellular calcium 334 and expresses functional NMDA re- ceptors 225. In immature neurons, the NR2B subunit of the NMDA receptor is shown to be accumulated in growth cones of axonal processes suggesting a role for this subunit in neurite outgrowth 114. Neurite outgrowth from iso- lated cells also referred to as neuritogenesis, plays an important role for cell- cell interactions. It is dependent on cytosolic Ca2+ and accordingly, activation of the NMDA receptor stimulates neuritogenesis 234,254. The NMDA receptor is also known to regulate neuritogenesis in primary cultures 36,61,213 and is believed to play an important role in neuritogenesis in the adult as well as in developing brain 213,214. However, it is noteworthy that the sigma 1 receptor, also in control of calcium , is previously described to induce neuritogenesis 286. NG108-15 (neuroblastoma x glioma) hybrid cells (obtained from ATCC, USA) were grown in 260 ml plastic culture flasks (75 cm2 bottom area; Nunc A/S, Roskilde, Denmark). Culture medium [D-MEM, supplemented with 10% foetal bovine serum (FBS) 100 U/ml G, 100 mg/ml strepto- mycin and HAT-supplement (hypoxanthine 5 mM, aminopterin 20 μM, thymidine 0.8 mM) (diluted 1:50)] was changed every second day. Conflu- ent grown cells were split 1:3 to fresh culture flasks. Cells at passages 5 – 25 were used.

Differentiation of NG108-15 The NG108-15 cells were allowed to grown to approximate 50% confluence, thereafter the cells were transferred to fresh 100 mm culture dishes (Sarstedts). The culture medium was changed to differentiating medium [D- MEM supplemented with 0.5% FBS, 1.5% DMSO 270, 100 U/ml penicillin G, 100 mg/ml streptomycin and HAT-supplement (diluted1:50)]. The cells were allowed to differentiate for 24 h, defined by growth characteristics ac- cording to Seidman and co-workers 270. Dishes were checked before treat- ment was started, using only dishes containing > 95% differentiated cells.

29 Membrane preparation Rat tissue membrane preparation for receptor binding (paper I & V) Membranes were prepared from the frontal cortex, a tissue rich in NMDA receptors and consists of circa 50% NR1/NR2A and 50% NR1/NR2B in adult animals 327 (paper I and V), cortex (paper V), the hippocampus (paper V) and the hypothalamus (paper V) tissues. The tissue was homogenized by means of a Teflon pestle homogenizer in ten volumes of ice-cold 50 mM Tris/acetate buffer supplemented with 0.32 M sucrose at pH 7.4. The ho- mogenate was centrifuged twice at 1,000 x g for ten minutes at 4 C. The supernatant was further centrifuged, at 17,000 x g, the resulting pellet was homogenized in 20 volumes of ice-cold 50 mM Tris/acetate buffer pH 7.0, and finally centrifuged three times at 50,000 x g for 30 minutes. The final pellet was re-suspended in ice-cold rat storage buffer [10 mM Tris/acetate (pH 7.0), 10% sucrose] and stored as 500 μl aliquots at -80 C. The protein content was determined by the method described by Lowry and co-workers 168.

Cell membrane preparation for receptor binding (paper II & III) The CHO-E2 cells were harvested after brief incubation in trypsine/EDTA at 37 C and centrifuged at 1,000 x g for 5 minutes. The supernatant was de- canted and the pellet was dissolved in cell buffer (50 mM Tris/HCl, pH 7.4). Centrifugation was performed three times and the final pellet was dissolved in a small amount of binding buffer, and homogenized in a polytron ho- mogenizer. More cell buffer was added to the homogenate, which was then centrifuged at 35,000 x g for 15 minutes. The pellet was dissolved in ice- cold cell storage buffer [10 mM Tris/HCl (pH 7.4), 10% sucrose], and the well-washed membranes were stored at -80 C. The protein content was measured by the method described by Lowry and co-workers 168.

Ligand binding 3H-ifenprodil receptor binding was conducted as described in paper I-III & V. In short, the rat frontal cortex homogenate was incubated with 3H- ifenprodil, unlabeled ifenprodil and neurosteroids for two hours at room temperature (paper I-II & V), 24 h at 4°C (paper II) or 30 min (paper II-III & V) in order to reach equilibrium. All mixing and dilution steps were per- formed in an automated robotic work station (Biomek® 2000). Incubation was terminated by rapid filtration under vacuum, using a Tomtec® 96 well

30 harvester and GF/A (paper I) or GF/B (paper II-III & V) filters. Filters were scintillated and measured for beta radiation in a TriLux® beta counter. Total specific binding, competition binding, and dissociation assays were carried out, using 0.6 nM (6.0 nM at 37°C; 1.5 nM in sigma receptor ex- periments) 3H-ifenprodil and in saturation analysis, concentrations were ranging between 0.01 and 32 nM (0.01 – 38 nM at 37°C). Non-specific bound 3H-ifenprodil was determined in the presence of 10 μM (100 μM at 37°C) unlabeled ifenprodil (NMDA receptor experiments) or 100 μM halop- eridol (sigma receptor experiments). Dissociation assays started after equi- librium was reached and induced by the addition of 100 μM (1 mM at 37°C) unlabeled ifenprodil. In paper I-III & V, trifluoroperazine was used to block all other sites for 3H-ifenprodil apart from the NR2B site. In paper V, 1.0 mM , 1.0 μM 272 and 50 nM GBR 12,935 (1-[2- (diphenylmethoxy) ethyl]-4-(3-phenylpropyl)-piperazine) 7,8,94 were used to block other sites (NR2B subunits, 1-receptors and piperazine acceptor sites respectively) for 3H-ifenprodil, when studying the sigma receptor binding of 3H-ifenprodil. In paper III, glutamate and glycine were added to the buffer in order to set the NMDA receptor in an activated state, this for the comparison to the data from intracellular calcium measurements.

Ligand binding calculations Saturation studies. The results from saturation experiments reveals the number of binding sites available (Bmax) for the radioligand, expressed as pmol bound radioligand per mg membrane proteins, and its affinity for the receptor, expressed in the apparent dissociation constant (Kd) in nanomolar. Saturation isotherms were created from three independent series of 3H-ifenprodil saturation binding experiments. The results from specific binding were analyzed using non- linear curve fitting and compared for the best fit (one or two binding sites), using the equations: Equation 1 [3 H-ifenprodil] B  B  eq max  3 for 1 binding site and K d [ H-ifenprodil] Equation 2 [3 H-ifenprodil] [3 H-ifenprodil] B  B   B  eq max 1st site  3 max 2nd site  3 K d 1st site [ H-ifenprodil] K d 2nd site [ H-ifenprodil]

3 for 2 binding sites, where Bmax is the maximum binding of H-ifenprodil and 3 Kd is the apparent dissociation constant of H-ifenprodil.

31 The saturation experiments were conducted by adding subsequently higher concentrations of 3H-ifenprodil (paper I & III) or by using the same concentrations of 3H-ifenprodil and adding subsequently higher concentra- tions of ifenprodil (paper II & V), adding the concentrations of labeled and non-labeled ifenprodil, a method possible since the receptor has equal affin- ity for labeled and non-labeled ifenprodil.

Competition studies The displacement studies of 3H-ifenprodil were performed with different concentrations of unlabeled ifenprodil (10 pM – 1 μM) (paper I-III & V) or haloperidol (100 pM – 100 μM) (paper II & V) or other tested substances (paper V). The results were analysed using non-linear curve fitting and com- pared for the best fit (one or two binding sites), using the equations: Equation 3

(B  B ) for 1 binding site and   sb nsb Beq Bnsb 110([competitor] - logIC50 )

Equation 4

(B  B ) (B  B ) B  B  f  sb nsb  f  sb nsb eq nsb 1st site ([competitor] - logIC50 ) 2nd site ([competitor] - logIC50 ) 110 1st site 110 2nd site

3 for 2 binding sites, where Bnsb is non-specifically bound H-ifenprodil, Bsb is 3 specifically bound H-ifenprodil, IC50 is the concentration of unlabeled ifen- prodil that causes 50% inhibition at the binding site and f is the fraction of binding sites (1st and 2nd sites refer to binding sites with high and low affinity 3 for H-ifenprodil). Ki values were calculated using the Cheng–Prusoff equa- tion 46: Equation 5

IC K  50 i [radioligand] 1  K d

th The Hill coefficient, nH, was introduced by Hill in the 1910 in an attempt to describe the oxygen binding to hemoglobin, which occurs with a positively cooperative binding. When nH = 1, the interaction between ligand and recep- tor obey the law of mass action, for a bimolecular reaction 118,163. A dis- 163 placement curve diverged from the sigmoid shape display a nH 1 . A Hill coefficient exceeding 1 indicates a positive cooperativity and when the Hill

32 coefficient is below 1, the mechanism involved can be negative cooperativ- ity, more than one binding site independently from each other or one binding site displaying different affinity states 65,163. The Hill coefficient was calcu- lated using the equation: Equation 6    (Bsb Bnsb ) Beq Bnsb  110( logIC50 - [competitor]) nH

The results from competition experiments reveal the specificity of ligands to the binding sites and present non-labeled ligands affinity as IC50 values re- calculated to Ki values (nanomolar). They also display the effects of allos- teric modulation more visually then the saturation experiment.

Dissociation studies

The experiment is primly determining the dissociation rate constant (Koff) (min-1), which is depending on the affinity for the substance to the receptor. Koff expresses the rate of the dissociation of the ligand-receptor complex. Its opposite is the association rate constant, Kon, which is the rate of forming the ligand-receptor complex. Together, they calculate the apparent dissociation constant, Kd: Equation 7 K  off K d Kon

The dissociation of 3H-ifenprodil from the NMDA receptor or the sigma receptor was studied at a fixed concentration of 3H-ifenprodil. The experi- ment began at equilibrium, reached after two hours at room temperature (paper I-II & V), 24 h at 4°C (paper II) or 30 min (paper II & III) and disso- ciation was initiated by the addition of excess unlabeled ifenprodil (paper I- III & V) or what is to be considered a large volume of buffer, 3000 μl (paper II). The results were analysed using non-linear curve fitting and compared for the best fit (one or two binding sites), using the equations: Equation 8

    K off t Bt Beq e for 1 binding site and

Equation 9

    for 2 binding sites, B  B  e Koff 1 t  B  e K off 2 t t eq 1st site eq 2nd site

33 where Bt is the specific binding at time point t and Beq is the specific binding st nd at equilibrium, Bt 2nd site is the plateau between the 1 and 2 binding sites and Koff 1 and Koff 2 are the dissociation rate constants.

Fluorescence measurements (paper III) 2+ Cell preparation for [Ca ]i measurements The cells were harvested after brief incubation in trypsine/EDTA at 37 C and centrifuged at 1000 x g for 5 minutes. The supernatant was decanted and the pellet was dissolved in a balanced salt solution (BSS). The BSS was used as a buffer with the purpose of keeping the cells viable and functional. The BSS contains 130 mM NaCl, 5.4 mM KCl, 2.0 mM CaCl2, 5.5 mM glucose and 10 mM HEPES, pH 7.3 305. Centrifugation was performed three times and the pellet was then dissolved in a small amount of BSS.

Loading of fluorescence Heat-activated cells were gently centrifuged (300 x g, 3 min), resuspended in 100 μL BSS supplemented with Fura-2 acetomethoxy ester (Fura-2 AM) (5.0 μM), 0.001% cremophore EL and probenecid (1.0 mM), and incubated for 45 minutes at 37°C. Extracellular Fura-2 AM was removed by dilution, gentle centrifugation (1000 x g for 5 minutes) and decantation, and the cells were resuspended in fresh BSS (50000 cells in 100 μL), now supplemented with 100 μM trifluoroperazine and distributed in 96-well black, round- 2+ bottom plates (NUNC, Copenhagen, Denmark) for immediate [Ca ]i meas- urement.

2+ [Ca ]i measurement Intracellular calcium measurements were run in a fluorescence image plate reader (BMG Polarstar, Labvision, Stockholm, Sweden) at 37°C. The cells were illuminated alternately at 340 and 380 nm. The emitted signals were recorded at 510 nm. The quotient of the emission generated from 380 nm divided by the emission generated from 340 nm was calculated. Baseline measurements were established and a mixture of glutamate and glycine (fi- nal concentrations of 100 μM and 10 μM, respectively, dissolved in BSS) was added to the well by a pump (100 mL min-1). This addition was per- formed in quadruplicate for every run and was used as an internal standard. The cells were pre-incubated, firstly with the neurosteroids and then with other substances, before measurement. The baseline was established and the glutamate and glycine solution was added. The peak signal was monitored

34 for 45 seconds per well. The peak counts were normalized to the internal standard and blank and were then translated into calcium ion concentrations.

Neurite measurement (paper IV) The neurite outgrowth was morphometrically analyzed as described by Wu et al. 334. The cells and the neurites were photographed (Minolta Z1 digital camera) through microscope (Olympus CK2) and 20x magnification. Each dish was photographed on three randomly picked spots at time zero (start of treatment), 24 h and 48 h after treatment. Photographs were transferred to, magnified and enhanced in Adobe Photoshop 3.0 (Adobe Systems Inc. 2005). The data recorded from each file include the number of neurites per cell, length of primary neurite, number of branch points per neurite and the size (thickness) of the neurites.

Data analyses Data is presented as mean ± S.E.M. The binding (paper I-III & V) and fluo- rescence (paper III) data were assembled, and analyzed using nonlinear and linear regression in Microsoft Excel (Microsoft Corp.1995-2003) and Prism 4.0 (GraphPad, 2003). The biphasic curves were compared with the mono- phasic curves using the sum of squares (partial F-test) and the biphasic curve was chosen when considered to be a statistically significant improvement. One-way ANOVA was followed by Dunnett's multiple-comparisons post- hoc test (binding and fluorescence data) or Fischer’s Protected Least Signifi- cant Difference (PLSD) post-hoc test (morphometry data). P < 0.05 was considered as the significant level.

35 Results and discussion

Optimizing the processes

Evaluation of the membrane preparation Stability of the test substances is vital to the process and the results. In order to assure that no sulfated neurosteroids were converted to non-sulfated neu- rosteroids, the membrane preparations were also conducted in the presence of estradiol (100 μM). Estradiol is a sulfatase inhibitor which pre- vents the transformation of sulfated neurosteroids to non-sulfated neuroster- oids 264. Since this membrane preparation yielded the same results as the membrane preparation without estradiol, estradiol was subsequently ex- cluded. The membrane preparations were also conducted in the presence of an enzyme inhibitor cocktail (Complete, Mini®), in order to evaluate receptor degradation during the homogenization procedure. Since there were no sig- nificant differences in total 3H-ifenprodil specific binding, enzyme inhibitors were subsequently excluded.

The discrimination of receptors (paper I-III & V) The binding of 3H-ifenprodil to the NMDA receptor (paper I-III & V) was discriminated to the NMDA by the co-incubation of trifluoroperazine block- ing all other sites 1,59,105,304. The low affinity NMDA receptor site was dis- criminated by the use of nanomolar levels of ifenprodil. The combination of GBR 12,909 and GBR 12,935, blocking the sigma 1 receptor and the piperazine acceptor site was a theoretical option for the study. The combina- tion was discarded in favour for the use of trifluoroperazine, which provides 304 the advantage to block 1-receptors as well, which is a target not only for ifenprodil but also for PS 70. The difference, in terms of receptor affinity, between 3H-ifenprodil and ifenprodil could be assumed to be neglected, why data from 3H-ifenprodil could be translated to non-labeled ifenprodil. The binding was best described using one-site binding equations (paper I- III & V) and the specific binding was kept above 91%. Ki- and Koff-

36 determinations are shown in Table 1. The nH showing the slope of the sig- moid displacement curve is close to -1 (perfect sigmoid shape), the evidence of one isolated binding site.

Table 1. Affinities of ifenprodil to the NMDA receptor. Ki-values are determined in 3 competition experiments, displacing H-ifenprodil with unlabeled ifenprodil, Koff- values are determined in kinetic experiments, using an excess of unlabeled ifen- prodil to induce the dissociation. Trifluoroperazine was used to block all but the NR2B site for ifenprodil. n.d. = not determined. * using glutamate activated recep- tors. † Unpublished data obtained during experiments for paper III. See Material & Methods for full description of experimental procedures.

-1 Tissue Temp. (°C) Ki (nM) nH Koff (min ) paper Rat the frontal cortex 25 4.8 ± 0.7 -0.92 0.20 ± 0.022 I Rat the frontal cortex 25 4.7 ± 0.5† -0.95 0.18 ± 0.018 V Rat the hippocampus 25 n.d. n.d. 0.21 ± 0.038 V Rat the hypothalamus 25 n.d. n.d. 0.16 ± 0.023 V Recombinant CHO 4 60.7 ± 7.5 -0.88 4.9 ± 0.52 II Recombinant CHO 25 57.2 ± 6.7 -0.91 5.1 ± 0.28 II Recombinant CHO 37 60.9 ± 5.8 -0.90 4.7 ± 0.34 II Recombinant CHO* 37 37.5 ± 5.5 -0.89 5.2 ± 0.013 III

The 3H-ifenprodil binding was limited to the sigma receptor by the pres- ence of 1 mM spermidine, blocking the NMDA receptor 268, 50 nM GBR 12,935, blocking the piperazine acceptor site 8 and 1 μM prazosin, blocking 272 the 1 receptor (paper V). It was best described using one-site binding equations and the specific binding was kept above 93%.

The effects of neurosteroids involving the NMDA receptor

Allosteric modulation of the 3H-ifenprodil binding by neurosteroids (paper I-III & V) The effects of PS and 35S on 3H-ifenprodil binding to the NMDA recep- tor (paper I-III & V) and ifenprodil (paper III) were studied at low neuros- teroid concentrations (0.1 – 100 nM), levels that possibly are more close to what could be considered as physiological 58,106,172,320 than experiments con- ducted with electrophysiological techniques using 100 μM of neurosteroids. The neurosteroids exerted no direct effects on neither the 3H-ifenprodil bind- ing, such as competitive displacement, nor on the calcium influx through the NMDA receptor on their own. Instead, the distinct effects appear to be

37 modulatory, shifting the binding properties for 3H-ifenprodil and the block- ing abilities for ifenprodil on the NMDA receptor in an allosteric manner. An allosteric modulating agent by binding to its site, changes the affinity for another ligand to its site. The changes in affinity is then best measured as changes in dissociation rate 135 of the receptor-radioligand complex, meas- ured as Koff, why the dissociation experiments is the best way of studying an allosteric modulation. Kon is depending only on concentration and keeping the concentration constant means that changes in the dissociation curve is 163 due to changes in the affinity / Koff . The possibility of the existence of two unique binding sites independent from each other or one binding site displaying different affinity states must be distinguished from negative cooperation. This is solved by performing to similar kinetic experiments: using “infinite dilution” only and compare this to “infinite dilution” plus excess of unlabeled ligand. The negative coopera- tive system will display an accelerating dissociation in the first case but not in the second one. When both experiments display the same dissociation profile, the system is composed of two binding sites or one binding sites with two different affinity states.

Different targets for PS and 35S (paper I-III) Many neurosteroids are suggested to have several different binding sites, not shared by all neurosteroids. PS and 35S are shown not share binding sites in the high micromolar concentrations, this in studies on apoptosis 231,315,333. In order to examine the possibility for the NMDA receptor of hosting two different high affinity sites for PS and 35S in the nanomolar concentra- tions, we investigated their pharmacology (paper I). The first indication on the existence of two sites was the observation that PS and S did not interact competitively. This conclusion is drawn from the observation that they did not significantly affect one another’s modulatory effect curves. It is indicating that PS positive and S negative effects on 3H-ifenprodil spe- cific binding are mediated through distinct sites. The existence of two sites is further supported by the findings that glutamate decreased the PS induced stimulatory effect by approximately 35% while glutamate had no effect on S mediated inhibition. These results show that the binding site for PS is sensitive to the state of the NMDA receptor (glutamate activated or not), an effect that may be mediated through an allosteric interaction between the glutamate binding site located on the NR2B subunit 156 and the putative high affinity PS site. It is consistent with the findings in electrophysiological stud- ies where the positive modulatory effect of PS is decreased during the acti- vated state of the receptor 125. When present alone, glutamate increased 3H- ifenprodil binding, an effect that has been reported by others 101. Though in contrast, no effect of the competitive glutamate antagonist APV on 3H- ifenprodil binding was observed in this study, as was reported in the study of

38 Grimwood and co-workers (2000). The differences in the results could be explained by the absence (our cell homogenate) respectively the presence (tissue homogenate) of endogenous glutamate, so the described effect of APV in fact is the result of absence of the stimulating glutamate effect. No effects of APV were to observe on the modulatory effects of neurosteroids on 3H-ifenprodil binding. Thus, the present results further underline earlier findings that ifenprodil binds with higher affinity to activated NMDA recep- tors (state-dependent binding) 144. The fact that glutamate had no effect on S induced changes of 3H-ifenprodil binding further support that its site is distinct from that of PS and suggests that the action of S is not de- pendent on the state of the NMDA receptor. Since neurosteroids appear to only have small structural differences (Figure 11) it may be difficult to be- lieve that they possess such great differences in action. But when the struc- tures are displayed in 3D (Figure 12) it can be more easily understood, where the double bond in steroid skeleton of PS forces a rather planar ap- pearance of the molecule and S is heavily bent between the A- and B- ring.

Figure 11. The 2D structures of PS (left) and S (right) show great similarities.

Figure 12. The 3D structures of PS (left) and S (right) reveal the big difference between the two neurosteroids. Illustration adopted from Weaver et al., 2000 315.

The effects on 3H-ifenprodil binding to the NMDA receptor, of ligands acting at other modulatory sites of the NMDA receptor, were also studied (paper I). Reduction of two cysteine residues in the NR1 subunit is shown to potentiate the NMDA receptor response 285. Such modification of the recep- tor complex, induced by the reducing agent dithiothreitol (DTT), had no

39 effect on the modulation of 3H-ifenprodil binding by PS or S. The ef- fects of the two neurosteroids were found to be unaffected in the presence of the co-agonist glycine which binding site resides exclusively on the NR1 subunit 119,151. Summing up the characteristics for the sites mediating the effects of PS and S on 3H-ifenprodil binding, it is clear that the phar- macology display several similarities with those observed in electrophysio- logical experiments. In such studies on chick spinal cord neurons and on NMDA receptors expressed in Xenopus laevis oocytes, PS and S were demonstrated also to act through different sites 232. Furthermore, the neuros- teroids are shown not to interact with the redox modulatory or the glycine site 231,232,333. The interaction between PS (at the low-affinity site) and gluta- mate on one hand and ifenprodil and glutamate on the other, have been sug- gested to be mediated through conformational changes of their binding sites located on the NR2B subunit 101,125. Since we here observe interactions com- parable to those found by these authors and no effect of the NR1 specific ligand glycine, it is tempting to speculate that the high-affinity site for PS, also is located on the NR2B subunit. As none of the allosteric ligands tested affected S mediated modula- tion of 3H-ifenprodil binding, it is likely that this neurosteroid act on a site not allosterically linked to any of those recognized by these ligands.

The neurosteroid effect curves are bell- or U-shaped (paper I-III) The high affinity sites for the neurosteroids on the NMDA receptor are only available in small concentrations interval, where the action is present. PS and 35S binding to the putative high affinity sites show opposite actions on the 3H-ifenprodil binding (paper I-III) and the ifenprodil calcium influx inhi- bition (paper III). Where PS increases the affinity for 3H-ifenprodil in a bell- shaped curve of action, 35S decreases it in a U-shaped curve of action. In the narrow window of high affinity action for neurosteroids, binding data indicates that PS or 35S both increase the number of accessible binding sites for 3H-ifenprodil and that the primary difference lays in the way they modulate the affinity for 3H-ifenprodil (paper I-III). Two other neurosteroids were investigated: we found that neither allo- pregnanolone sulfate (ALLOPREGS; 35S) nor DHEAS changed the number of binding sites for 3H-ifenprodil. Nor did they affect the affinity for the radioligands in saturation and dissociation experiments. Further experi- ments could however bring more light to their modulatory activities, since competition data indicates that certain concentrations are able to change the curve fit into a two-site fit, 10 nM DHEAS slightly lowered the IC50 for ifenprodil 136. Interestingly, where low doses of PS induced improvement of cognitive functions there were also a description of a biphasic dose-response curve 80 and the anti- effect of DHEAS was limited to a bell-shaped dose

40 interval 255. Furthermore, biphasic effects have also been demonstrated to occur for other modulating factors acting on the NMDA receptor. For exam- ple, low doses of cations or polyamines stimulate binding of the non- competitive channel blocker 3H-MK-801 and the NMDA site selective an- tagonist 3H-CGP 39,653 to rat brain membranes while high doses reduce such binding 212. A similar effect has also been described for polyamines in studies on 3H-MK-801 binding to porcine hippocampal membranes 122. The inorganic divalent cation Zn2+ has also been demonstrated to display bell- shaped dose-response curves in desensitization experiments measuring cal- cium current in HEK-293 cells expressing NR1/NR2A receptors 338. This phenomenon may reflect high and low affinity states of the same binding site or distinct sites with different affinities for the modulators.

Temperature involvement (paper II) Ifenprodil binding is sensitive to temperature. In assays run at 4°C, ifen- prodil binds approximately equally to the ifenprodil site on the NR2B sub- unit and to the sigma receptor 110. At 37°C, more than 90% of the ifenprodil binding is to the sigma receptor 108. With the temperature impact on ifen- prodil selectivity in mind, the issue had to be investigated thoroughly: since the discovery of the neurosteroid modulatory effects on 3H-ifenprodil bind- ing on rat the frontal cortex membranes was made at 25°C and since the functional experiments had to be run at 37°C on recombinant NMDA recep- tors, the data would have been hard to translate. The recombinant NR1/NR2B receptors on CHO-E2 membranes were subsequently ran at 4°C, room temperature (25°C) and body temperature (37°C) (paper II), evaluating the binding characteristics for 3H-ifenprodil. In agreement with the findings of Hashimoto and London (1993) and Hashimoto and co-workers (1994) 108,110 3 , the total available NR2B sites (Bmax) for H-ifenprodil was signifi- cantly higher at 4°C compared to 37°C. Compared to 4°C, there was a ten- dency towards less number available at 25°C, but not statistically significant. More important was the finding that the dissociation constant rate Koff for 3H-ifenprodil to the NR2B subunit was independent of the temperature. The apparent affinity increased at 4°C compared to 37°C displaying a lowered Kd. Since Koff was unaltered, the change in Kd has to depend on the associa- tion rate (see Equation 7). This fact is supported by thermodynamic laws, 3 when in the assay, the concentration of H-ifenprodil was far below the Kd, the association is directed by the concentration of receptors and ligands and the temperature 163. The more heat, the more energy, the faster association and Kon are increased, resulting in a lowered Kd. Kon and Koff are generally targets for changes in temperature 10,33,173. But temperature does not always dictate the kinetics parameters 145,162,316. Haloperidol binding at dopaminergic receptors is non-sensitive to alterations in the temperature 146 and ion channel kinetics often shown temperature independence 33. Since haloperidol and

41 ifenprodil shown similarities in kinetics and binding properties to the NMDA receptor, it seems reasonable that the dissociation could be insensi- tive to temperature.

Allosteric & direct effects of neurosteroids (paper I-III & V) The NMDA receptor is modulated and fine-tuned, not only by single interac- tions with ligands, but by complex interactions between protons, poly- amines, neurosteroids and ifenprodil. The protons are believed to play a cen- tral role in the regulation. Protons induce a tonic inhibition of the receptor at physiological pH. The inhibition is about 50% in the normal state 289,297. Polyamines are relieving this inhibition, leading to a potentiation of the re- ceptor activity, and ifenprodil is somewhat counteracting the polyamine ac- tion, increasing the proton inhibition 167,179,210,229,298. The present findings are examples of those complex interactions. Neuros- teroids in nanomolar concentrations do not have any direct influence on the intracellular calcium. Neither did pre-incubation with the neurosteroids alter the response of the NMDA receptor when stimulated with glutamate and glycine. Instead they appeared able to modulate the calcium influx via inter- actions with ifenprodil at low concentrations. When glutamate and glycine stimulated and ifenprodil inhibited the receptors, the inhibiting action from ifenprodil was changed when pre-incubated with PS or 35S. Pre- incubation with PS caused increased inhibition and pre-incubation with 35S gave less inhibition. This suggests that PS increase the ifenprodil affinity and thereby its inhibiting effect, whereas 35S produces an oppo- site action (paper III). The neurite outgrowth and morphology in NG108-15 cells, is a process dependent on intracellular calcium. Experiments presented in this thesis con- firmed that nanomolar concentrations of S induce effects only via ifen- prodil modulation, whereas nanomolar concentrations of PS exerted a somewhat more complex pattern of effects (paper IV). Neurosteroids are previously shown to regulate the morphology of astrocytes 64. The changes in morphologic characteristics in the neurite outgrowth, due to NMDA re- ceptor stimulation and modulation of this were studied in differentiated NG108-15 cells (Figure 13a). Stimulation with glutamate and glycine in- duced the neurites to grow thicker, longer and produce more branch point (Figure 13b). Branch points are nodes where neurites are connected or the possibility for a new neurite to launch. Stimulation with glutamate and gly- cine and inhibition with ifenprodil reduced the stimulatory outgrowth effect (Figure 13c). The combined addition of ifenprodil and PS to the glutamate and glycine stimulation was not different from ifenprodil blockade to gluta- mate and glycine stimulation after 24 h (no figure) but after 48 h the neurites were fragmented and the cells were detached from the surface (Figure 13d). The combined addition of ifenprodil and 35S to the glutamate and glycine

42 reduced the ifenprodil blockade after 24 h (Figure 13e) but that effect was not to be seen after 48 h (Figure 13f).

(a) (b) (c)

(d) (e) (f)

Figure 13. The differentiated NG108-15 cells after 48 h of absence or presence of treatment [except from (e) which is after 24 h]. (a) non-treated, (b) glutamate and glycine treatment, (c) glutamate and glycine treatment combined with ifenprodil, (d) glutamate and glycine treatment combined with ifenprodil and PS, (e & f) glutamate and glycine treatment combined with ifenprodil and 35S.

Since the outgrowth itself is a slow process, the neurites were examined after 24 h and 48 h. The experiment is troubled by the time span and cannot for certain exclude genomic effects from the neurosteroids. Indeed, the ef- fects of PS alone, is as likely to be genomic as non-genomic, whereas the effects seen in the presence of S at least partly may be due to interac- tion with ifenprodil since S attenuate the effects of ifenprodil after 24 h, but not after 48 h (paper IV). There are recordings that there are direct interactions between the NMDA receptor and neurosteroids, not involving other NMDA receptor agents. This is only documented for neurosteroids at high concentrations. The specific target for neurosteroids on the NMDA receptor is called steroid modulatory domain (SMD). Activation via SMD requires high concentrations of the neurosteroids (approx. 100 μM) 132, why SMD could be considered as a low affinity site. Electrophysiological experiments using high micromolar con- centrations of PS and S define PS as a positive modulator and S as a negative modulator of the NMDA receptor 42,125,174, effects which possi- ble originate from the interaction between the neurosteroids and SMD. These

43 effects are described to take place in the presences as well as in the absence of glutamate activation. The target or targets for lower concentrations of neurosteroids on the NMDA receptor remain(s) undefined, but may well be high affinity site(s), completely different from SMD, as the effects seen at much higher concen- trations in electrophysiological experiments are independent of other ligands. This is illustrated in a model (Figure 14) where one NR1 and one NR2B with these and other known binding sites are shown, together with their in- teractions. The existence of both high and low affinity binding sites for neu- rosteroids on the NMDA receptor complex, could explain the difference between the direct effects seen in electrophysiological experiments and the modulatory action on cognitive functions 79,80,180,181,193 as well as the modulation of 3H-ifenprodil binding and ifenprodil inhibition, as seen in paper I-III & V.

Figure 14. Schematic view of the two subunit types forming the NMDA receptor with binding sites and the results of binding to those sites. The ion pore is blocked by Mg2+ in the non-activated state. Glutamate and glycine are the agonist and co- agonist inducing the gate opening. Their binding also promotes the binding of the channel blocker MK-801. Glutamate and ifenprodil increases each others binding. Protons inhibit the gate opening and micromolar concentrations of neurosteroids at SMD can either inhibit or promote the gate opening. Reduction or oxidation of the sulfur bonds inhibit or amplify the amplifying effect of Zn2+ on the inhibiting effect of protons. Polyamines and ifenprodil bind to separate targets which interfere with each other causing decreased binding. Ifenprodil, as well as haloperidol, increase the proton inhibition, whereas polyamines relieve the proton inhibition. Neurosteroids in nanomolar concentrations seems to have different targets, one acting positively on the ifenprodil binding and one negatively.

44 The effects of neurosteroids involving the sigma receptor (paper IV-V) The sigma receptor is shown to bind neurosteroids. This raised an interest to investigate a possible interaction between ifenprodil and the neurosteroids at the sigma receptor. PS, 35S and DHEAS at micromolar concentrations all displaced 3H-ifenprodil from the sigma 1 and the sigma 2 receptor (paper V). Previously, it is shown that PS, DHEAS and PROG have affinity to the sigma 1 receptor and to the sigma 2 receptor 205. All these three neuroster- oids display a planar structure due to the unsaturation in the A-rings. PS and DHEAS act as agonists and PROG acts as antagonist at the sigma 1 receptor 20,204. An interesting difference is that the agonists are sulfated neurosteroids and the antagonist is not. Those sigma 1 receptor agonistic properties of DHEAS have been dem- onstrated to counteract the memory deficit effect of MK-801 186 and inhibit persistent Na+ currents 47. Concerning PS, its binding to and activation of a pre-synaptic sigma 1-like receptor in male Sprague-Dawley rat the hippo- campus 70,267 and cortex 70, induce the NMDA receptor facilitated excitatory post-synaptic current (EPSC). PREG is suggested to induce glutamate re- lease via the sigma 1 receptor 200. The effects seen on neurite outgrowth and morphology in NG108-15 cells were addressed to be mediated by interactions with the NMDA receptor (paper IV). However, it could as well be mediated by the sigma receptor, which is present in the cell line 90,331. Talking against it, is the fact that it is believed that sigma receptors mainly exists in the endoplasmatic reticulum and not in the membrane 113. But on the other hand, the use of the sigma receptor blocker trifluoroperazine, as in receptor binding and fluorescence experiments, was not possible due to unwanted reactions as ceased cell divi- sion, cell processes, as neurite growth, terminated and the cells assumpted to spherical shape and ceased cell attachment to the surface. The assumption to spherical shape symptom is reported to be a sigma mediated effect in C6 glioma cells 329,330. It is possible to speculate that the massive concentration of trifluoroperazine caused the cell detachment by interactions with sigma receptors. Then, it could be the same effect produced by the neurosteroids, mainly PS. It is strictly hypothetic and further studies are needed in order to shed light on the issue.

Influences of AAS on neurosteroid effects (paper V) The AAS nandrolone decanoate treatment has been reported to demonstrate central side effects. The effects are rapid, occurring within minutes after administration. This is considered to be mediated by non-classical mecha-

45 nism. Possible explanations could be interference with neurosteroids, with the or formation of neurosteroids. In fact, the AASs danazol and stanozolol have been shown to have affinity for non-androgenic receptors 31. Since AAS act via classical (genomic) mechanisms as well, it is not unlikely that AAS could have influence on neurosteroids targets after chronic admini- stration. Previous studies have shown alterations of the dopaminergic 27,148, the serotonergic 147, the GABAergic 29,30 and the glutaminergic systems 157,158 after chronic or sub-chronic treatment with nandrolone decanoate. The theory of the interference between AAS and neurosteroids was stud- ied in paper V. The glutaminergic system was previously studied on the mRNA level 157,158. This investigation of the NMDA receptor was conducted at protein level. The ifenprodil binding site on the NR2B subunit after chronic nandrolone decanoate treatment was studied, not only looking at the frontal cortex, but also at the hippocampus and the hypothalamus. There was no up- or down-regulation of the receptor, measured as 3H-ifenprodil binding to the NR2B subunit, as result of nandrolone decanoate administration. No difference was recorded in 3H-ifenprodil binding kinetics to the NR2B sub- unit between the AAS group and the control group. This was the case in all of the three regions. It is in agreement with previous studies of mRNA lev- els, except from the the hypothalamus where an up-regulation of the mRNA for NR2B was seen 157. A change in mRNA levels does however not neces- sarily correspond to altered protein expression. The kinetics of 3H-ifenprodil at the NMDA receptor was also unaltered of AAS administration, as was the modulatory effects of PS and 35S at nanomolar concentrations. The conclusions drawn from those findings there- fore became that chronic nandrolone decanoate treatment neither affect the neurosteroid and ifenprodil binding sites nor the NR2B subunits in the stud- ied regions. Turning the attention to another ifenprodil and neurosteroid target, the sigma receptor, the outcome was different. The chronic treatment with nan- drolone decanoate changed the target for the neurosteroids on the sigma 1 receptor, decreasing their ability to displace 3H-ifenprodil from the sigma 1 receptor. The sigma 2 receptor was unaffected. Interestingly, the results also proved evidence that 3H-ifenprodil and neurosteroids do not share binding site on the sigma 1 receptor, but that the neurosteroid binding interferes with and blocks the 3H-ifenprodil binding, since 3H-ifenprodil, ifenprodil, halop- eridol, trifluoroperazine and (+)-SKF 10,047 binding to the sigma 1 receptor was unaltered by the AAS treatment. Another steroid overload that affects the sigma receptor function in the brain is 21, why it seems rea- sonable that AAS could target the sigma receptor. This is probably a result of genomic interaction with the complex of the receptor and nandrolone, since the change is still there after de- capitation, dissection and membrane preparation. It could also be the conse- quence of high concentrations around the receptor, which has changed its

46 nature due to input from the immediate environment. Whether such effect can persist to the in vitro assay has to be further investigated. These experiments leave a gap to be filled, since it does not include direct and rapid effect of nandrolone or nandrolone decanoate similar to the allos- teric effects produced by the neurosteroids. It does neither tell about interac- tions nor competitions between the AAS and the neurosteroids.

Imagining the use for neurosteroids

Neurosteroids in combating drug abuse The NMDA receptor, and in particular the NR2B subunit, is believed to be involved in the development as well as the maintenance of drug and dependence 28,215. Antagonists to the NMDA receptor are proposed to be effective in drug addiction treatment, diminishing and attenuate both the responses from drug stimuli and the withdrawal symptoms 28. They also seems to counteract the physical dependence, sensitization and tolerance to opiates 299. Concerning the reported side effects from NMDA antagonists at therapeutic concentrations, the use of fine-tuning allosteric modulators as neurosteroids may replace or at least decrease the required doses of the an- tagonists, making the doses required tolerable for human therapeutic use. Neurosteroids possessing antagonistic properties at the sigma 1 receptor could be useful in the treatment of addiction to or co- caine. The sigma 1 receptor is a target for methamphetamine 251 and the sigma 1 receptor is up-regulated in the whole brain by 166. This is suggesting a possible therapeutic role for sigma ligands in cocaine 166 and methamphetamine dependence treatment, for instance neurosteroids.

Antidepressive and anxiolytic use of neurosteroids Several studies have provided evidence for the involvement of the sigma 1 receptor in depression 182,238,278 and the anti-depressive treatment 66,277,306,309. There is upcoming evidence for that the anti-depressive effects of SSRI, like fluoxetine (Fontex®, Prozac®) 263 and SNRI, like venlafaxin (Efexor®) 66 are mediated by sigma 1 receptor interaction and that neurosteroids can be used in treatment of depression due to sigma 1 receptor interactions 189,191. Sigma 2 receptor ligands produce potent anxiolytic effect, which probably but not certainly is sigma 2 receptor mediated, since the selectivity between the two sigma subtypes not is complete 262. They have been shown to induce apoptosis in cancer cells, why those ligands could be of use in anti-cancer therapy 141. Cancer might be a disease followed by anxiety, why anxiolytic properties of the anti-cancer drug could be of use.

47 The abuse of AAS is often accompanied by depression, anxiety and ag- gressive behavior. Chronic AAS treatment has been shown to induce anxiety 29,30 by altering the GABAA receptor in the cortex , but as mentioned above, these symptoms are also induced by the sigma receptors. Thus, those symp- toms could be the result of altered sigma 1 receptors or altered levels of neu- rosteroids, induced by the AAS. Therefore, the hypothesis could be formed, that chronic AAS administration is followed by losses in sigma receptor function. This is expressed as above mentioned symptoms. Therefore, before it has been fully examined, the influences of AAS on the sigma receptor cannot be ruled out. Thus, neurosteroids seems to be potential agents in the treatment of de- pression and anxiety, two often accompanied syndromes that causes big economical losses to the society and personal suffering.

Neuroprotective use of neurosteroids The need for neuroprotection is present in diseases as Alzheimer’s disease, Parkinson’s disease, stroke, and partum hypoxia injuries. Neurosteroids and NMDA receptor antagonists are both groups of sub- stances with neuroprotective properties. The affinity for several other recep- tors has limited the clinical use of competitive and non-competitive drugs, e.g. ifenprodil. Ifenprodil was once first defined as an anti-hypertensive drug 104,170, then thought of as a neuroprotective drug 96,265,272 assigned to its NMDA antagonistic effects. The non-tolerable side-effects thwarted the plans. The family of allosteric modulators is attractive as therapeutic agents with unique advantages to conventional agonists and antagonists due to their state-dependent action. In this context the effect of PS to stimulate the in- hibitory effect of ifenprodil effect could be of clinical interest, since a main- tained effect at decreased doses of the NR2B antagonist may help to reduce its side effects. Still, ifenprodil could face limited clinical usefulness, since NMDA receptor antagonists display side effects connected to the NMDA effect. The side effects include nausea, vomiting, psychosis and memory impairment. However, the weak or partial NMDA receptor antagonist me- mantine seems to be well-tolerable and still efficient in neuroprotection 88. And there have been/are derivates of ifenprodil in clinical trials for neuro- protection, lacking some of ifenprodil’s side-effects 45. Whether it is the lack of other targets for memantine or the weak effect on the NMDA receptor that makes it well-tolerable is not known, but if the tolerance is due to weak NMDA effect, it could mean that ifenprodil would be tolerated clinically if combined with 35S, attenuating its NMDA antagonistic effect. The sulfated neurosteroids themselves are metabolites of respective non- sulfated neurosteroid. Desulfatation is the reversed reaction, where non- sulfated neurosteroids are formed from sulfated neurosteroids. It does take

48 place and if it is not saturable, treatment with sulfated neurosteroids will result in elevation of non-sulfated neurosteroids and the risk for non-desired effects increases. Especially in the case of PS, which is converted to PREG, from which several, almost every neurosteroid can be formed (Figure 3). Another question mark using highly water soluble sulfated neurosteroids as drugs has to be raised, namely whether they pass the blood brain barrier and are able to reach their targets.

49 Conclusions

This thesis reports the findings that neurosteroids in physiological concentra- tions have distinct effects on the NMDA. Neurosteroids in therapeutic con- centrations have affinity to sigma receptors. It also reports changes of the sigma receptor, but not the NMDA receptor, from chronic treatment with the AAS nandrolone decanoate.

 The neurosteroids PS and 35S in nanomolar concentration medi- ate their influences on the NMDA receptor via allosteric interac- tions, not via competitive binding. The interactions are robust to changes in the temperature.

 The modulatory effects were seen within narrow windows of nano- molar concentrations (0.1 – 10 respectively 1 – 100 nM) of the neu- rosteroids suggesting that they act through binding sites with higher affinity for the neurosteroids compared to those mediating the action on NMDA receptors observed in electrophysiological studies. At least two different sites for neurosteroids are involved, which could be classified as high affinity sites.

 The neurosteroids in those nanomolar concentrations possess the ability to change the kinetics for 3H-ifenprodil binding to the NR2B subunit of the NMDA receptor, as well as the calcium influx inhibi- tory effect of ifenprodil. Whether this is the result of conformal changes of the known ifenprodil binding site or if the neurosteroids unveil other sites for ifenprodil remains to be further elucidated.

 The chronic treatment with the AAS nandrolone decanoate genomi- cally altered the binding for the neurosteroids, but not for 3H- ifenprodil, to the sigma 1 receptor. The conditions at the NMDA re- ceptor were unchanged for both the neurosteroids and 3H-ifenprodil.

 The neurosteroids did not displace all 3H-ifenprodil from the sigma 1 receptor. Together with the above mentioned, this indicates that 3H- ifenprodil and neurosteroids do not share the same binding site on the sigma 1 receptor. The partial ability to displace 3H-ifenprodil from the sigma 1 receptor could possibly be explained by the local-

50 ization of the two binding sites, perhaps in a similar way like ifen- prodil and polyamines do not share binding site on the NR2B sub- unit, but interfere in each others binding, probably due to very closely located binding sites.

This thesis concludes that there are two high affinity targets for neuroster- oids on the NMDA receptor. They are not previously described and they are not shared by other common NMDA receptor ligands or neurosteroids in high concentrations. We speculate that the structure, especially the possibil- ity or impossibility to bend the A-ring, is important for the effect of the neu- rosteroid. Together, this could be a valuable pathway for explaining effects of neurosteroids in vivo. These targets on the NMDA receptor are robust to chronic and supratherapeutic doses of the AAS nandrolone decanoate, doses that are comparable to those used by AAS abusers. The treatment altered the sigma target for the neurosteroids, why we can speculate that the depressions associated with AAS abuse could be the result of sigma receptor changes.

51 Future perspective

The interactions between ifenprodil, haloperidol and neurosteroids at the sigma receptor need further investigation, and their binding characteristics need to be elucidated. Neurosteroids and ifenprodil both interact on both the NMDA receptor and the sigma 1 receptor, which receptors are linked together and both con- trol intracellular calcium. The effects of neurosteroids on ifenprodil actions could be tested in an in vitro model, both receptors non-blocked, providing data on how the complete system controls the intracellular calcium. The NMDA receptors are at tonic proton inhibition at physiological con- ditions. It is believed that ifenprodil increases the proton inhibition and that polyamines relieve the same. It has also been proposed that neurosteroids also are involved in regulating the proton inhibition. Taken together, the effects of neurosteroids at different pHs could not only add knowledge to the nature of the neurosteroids and the NMDA receptor, but also about diseases believed to be proton mediated, e.g. epilepsy 216,224,318. Neurosteroids, and especially the balance between neurosteroids, are shown to be involved in epilepsy 83,84,116. Phosphorylation of the NR2 subunits seems to be of great importance for its function 155 and the NR2B subunit is the most prominent phosphorylated NMDA receptor protein 198. NMDA receptors being phos- phorylated on the NR2B subunit, by ERK dependent reactions, cause epi- lepsy when activated 216. There has recently been described that there is a third NMDA receptor subunit, the NR3 73,74,219 as well as non-classically composed NMDA recep- tors. Non-classically composed NMDA receptors are composed of other combinations of subunits than two similar NR1s and two similar NR2s, e.g. two NR1s, one NR2A and one NR2B 274. The incorporation of NR3 subunits would also create a non-classically composed NMDA receptor. It would be of interest to investigate whether the NR3 subunit has binding sites for neu- rosteroids, and it would also be interesting to study and characterize the non- classically composed NMDA receptor, in terms of responses to and behav- iour in the presence of neurosteroids. The lack of information of the NR3 subunit, binding sites and expression in the rat brain, makes it impossible to rule of that some of the data presented in this thesis actually derives from NR3 binding. Indeed, recombinant expressed non-classically composed NMDA receptors could be a useful tool in this question.

52 The allosteric effects on the ifenprodil binding site on the NMDA recep- tor could very well be unique, but imagining a conformal change in the bind- ing site, this change could perhaps be present in a nearby situated site, namely the polyamine site. The ifenprodil binding site on the NMDA recep- tor is situated nearby one of the polyamine sites, so close that binding to one of the sites interfere with binding to the other. This once lead to the misun- derstanding that ifenprodil was a polyamine antagonist, since polyamines apparently displaced radiolabeled ifenprodil 268. This could be elucidated using a radiolabeled polyamine, e.g. 3H-spermidine. Lots of experiments that have been undertaken, considering neurosteroid in vitro assays, have been performed using solvents like ethanol or DMSO. In the initial phase of our experiments, we planned to run both sulfated and non-sulfated neurosteroids. However, the solvents necessary to dissolve the non-sulfated neurosteroids severely changed the binding results for the sul- fated neurosteroids compared to running the assay in just water-based buffer, why we focused on the sulfated neurosteroids from the beginning. It would be a convenient continuation to optimize the present assay for solvents enough to keep non-sulfated neurosteroids in solution, to test those neuros- teroids. It would be particularly interesting to compare PREG and preg- nanolone with PS and 35S in this new environment and also to compare those results to the results presented in this thesis. Another issue, concerning the binding of neurosteroids, is the problem of measuring the direct binding, which seems impossible to membrane struc- tures. This has been explained with the model (Figure 15) where the neuros- teroid bind to its target, the target then alters configuration as a result of the interaction. The configuration alteration not only induces an allosteric modu- lation of other ligands, e.g. ifenprodil, but also ejects the neurosteroid itself. That means that the time of neurosteroid binding is too short to be measured with conventional binding assays. One way of dealing with and detecting this kind of binding could be to monitor the binding in real time.

Figure 15. A hypothetical and schematic model of the binding of neurosteroids to membrane targets, their allosteric effect on ifenprodil and the ejection of the neuros- teroids.

53 Populärvetenskaplig sammanfattning

Neurosteroider kallas de hormoner av steroidtyp som kan bildas i och ger snabba effekter i hjärnan. De är viktiga för processer som minne och inlär- ning, neuroprotektion, sinnesstämning samt neuronal placticitet. Neurosteroider bildas från kolesterol eller från andra steroidhormoner, som t.ex. könshormonerna. Effekterna medieras via interaktioner med mem- branbundna receptorer, såsom NMDA-, sigma- och GABA-receptorer. Me- kanismen som ger snabba effekter skiljer sig från den mer långsamma klas- siska där steroider utövar sin effekt genom att verka på genomet. Syftet med denna avhandling var att identifiera och karaktärisera de mo- lekylära mekanismerna för nanomolära koncentrationer av neurosteroider på NMDA receptorn, en koncentration som kan antas vara fysiologisk. Resultaten visar att neurosteroidererna pregnenolonsulfat (PS) och preg- nanolonsulfat (35S) förändrar bindningen av ifenprodil indirekt, alloster- iskt, till NMDA-receptorn, genom att binda till separate, unika ställen på NR2B-subenheten. Effekterna verkar vara temperaturoberoende och bekräf- tasdes vidare in funktionell kalciummätningsstudie. En andra funktionell studie visade att PS och 35S påverkar glutamatstimulerad neurittillväxt I NG108-15-celler. Anabola androgena steroider (AAS) inbegriper testosteron och testoste- ronliknande steroider. De har en begränsad klinisk användning, och då mest som ersättning för otillräcklig egenproduktion. Den stora användningen av AAS finner vi idag dels bland idrottsutövare och dels bland kriminella ele- ment. Missbruk av AAS kan ge kraftiga psykiska biverkningar, som ångest, depression, aggresivitet och förlorad impulskontroll. Eftersom neurosteroi- der är inblandade i processer som involverar humöret, kan de misstänkas att AAS och neurosteroider påverkar varandra i hjärnan. Kronisk administre- ring av AAS:en nandrolondekanoat förändrade dock inte de allosteriska ef- fekterna som PS och 35S utövar på NMDA-receptorn, men behandlingen förändrade affiniteten för PS, 35S och dehydroepiandosteronsulfat (DHEAS) till sigma-1-receptorn. Dessa resultat visade också att neurosteroi- derna konkurrerar med 3H-ifenprodil om bindningen till sigma-1- och sigma- 2-receptorer, utan att direkt dela bindingsställe på sigma-1-receptorn. Den minskade affiniteten för neurosteroiderna till sigma-1-receptorn skulle kunna förklara de depressiva symptom som ses vid AAS-missbruk. NMDA-receptorsystemet är djupt inblandat i neurodegeneration, celldöd och neurotoxicitet. Genom att blockera NMDA-receptorer kan neuronala

54 skador, t.ex. efter stroke eller vid Alzheimers sjukdom, begränsas eller hävas. Blockad av NMDA-receptorn är dock förknippat med mycket biverkningar, som i fallet med ifenprodil. Dessa fynd att neurosteroider och ifenprodil interagerar med varandra på NMDA-receptorn kan öppna för möjligheten att utnyttja en neuroprotektiv synergism, och därigenom komma ned på tolererbara doser för människa.

55 Acknowledgements

This work was carried out at the Department of Pharmaceutical Bio- science, Division of Research on Biological Drug Dependence, Faculty of Pharmacy, Uppsala University. The work was financially supported by The Swedish Research Council and Gunvor och Josef Anérs stiftelse. Travel grants from The Swedish Academy of Pharmaceutical Science (IFs stiftelse and CD Carlssons stiftelse).

I would like to express my sincere gratitude to all of you who has con- tributed to this thesis and I would especially like to thank:

Professor Fred Nyberg, Associate professor Pierre my supervisor, for giving me Le Grevès, my supervisor, for the opportunity to become a introducing me to research, PhD student in your group, for your invaluable contribution to giving me the opportunity to the projects, for all great scien- travel and for supporting and tific discussions and for always believing in me. keeping the greater picture in mind.

Dr. Per-Anders Frändberg, my co-author, for teaching me sci- entific skills, for always being there for a discussion of methods or results and for good advising.

Dr. Qin Zhou, co-author, for your excellent skills with animals and for teaching me the brain dissection techniques.

Carolina Birgner, my roommate, for pleasant tea drinking, for discussing everything and for making these years endurable.

Martin Elfverson, my co-author, for all nice discussions, co- working and for being a friend.

56 Uwe Rossbach, my colleague, for being a bench-mark in the laboratory and a friend.

Mrs. Britt-Marie Johansson, for always keeping the laboratory up and running, no matter who tried to ruin it. You made my work a lot easier!

Magnus Jansson, for sharing my interest in Formula 1 and for using his magic to repeatedly fix my computer.

Associate professor Mathias Hallberg, for collaboration during the “LOA-days”.

Past and present members of the group, Milad, Madeleine, Kristina, Jenny, Alfhild and Nasim for creating the atmosphere.

Past and present members of the staff, Hans Darberg, for manu- facturing everything I ever needed, Agneta Bergström for all ad- ministration, Marianne Danersund for managing all travels and Kjell Åkerlund for all support.

Past and present colleagues at BMC, Johan Alsiö, Mats Nilsson, Sadia Oreland, Ulrike Garscha, Henrik Alm, Anna-Malin Hermansson and Erik Hedner, for nice collaborations and chats.

Stefan Fritz, for being a good friend, always there ready to dis- cuss the life, the meaning of life and the benefit of life.

Mats Björving & Anders Österholm, for everything we share and for always being there.

Niklas Kåla Pettersson, for joining in the gym, the bear drink- ing in the sauna and mending cars.

My other friends, no one mentioned, no one forgotten.

My parents, Bertil and Christina, my grandparents Wivan and Yngve and my brother Martin, for there love and support during all these years.

Finally, to my family, Arianne and Emma, for your patience and love. You are the true sunshine of my life.

57 References

1. Abreu P, Sugden D: Characterization of binding sites for [3H]-DTG, a selective sigma receptor ligand, in the sheep pineal gland. Biochem Biophys Res Com- mun 171:875-881, 1990 2. Achermann JC, Silverman BL: Dehydroepiandrosterone replacement for pa- tients with adrenal insufficiency. Lancet 357:1381-1382, 2001 3. Agis-Balboa RC, Pinna G, Pibiri F, et al: Down-regulation of neurosteroid biosynthesis in corticolimbic circuits mediates social isolation-induced behavior in mice. Proc Natl Acad Sci U S A 104:18736-18741, 2007 4. Aitken C, Delalande C, Stanton K: Pumping iron, risking infection? Exposure to hepatitis C, hepatitis B and HIV among anabolic-androgenic steroid injectors in Victoria, Australia. Drug Alcohol Depend 65:303-308, 2002 5. Aizenman E, Lipton SA, Loring RH: Selective modulation of NMDA responses by reduction and oxidation. Neuron 2:1257-1263, 1989 6. Alonso G, Phan V, Guillemain I, et al: Immunocytochemical localization of the sigma(1) receptor in the adult rat central nervous system. Neuroscience 97:155- 170, 2000 7. Andersen PH: Biochemical and pharmacological characterization of [3H]GBR 12935 binding in vitro to rat striatal membranes: labeling of the dopamine up- take complex. J Neurochem 48:1887-1896, 1987 8. Andersen PH, Jansen JA, Nielsen EB: [3H]GBR 12935 binding in vivo in mouse brain: labelling of a piperazine acceptor site. Eur J Pharmacol 144:1-6, 1987 9. Areosa SA, Sherriff F, McShane R: Memantine for dementia. Cochrane Data- base Syst Rev:CD003154, 2005 10. Arya DP, Bruice TC: Triple-helix formation of DNA oligomers with methyl- thiourea-linked nucleosides (DNmt): a kinetic and thermodynamic analysis. Proc Natl Acad Sci U S A 96:4384-4389, 1999 11. Barann M, Bonisch H, Urban BW, et al: Inhibition of 5-HT3 receptor cation channels by ifenprodil in excised patches of N1E-115 cells. Naunyn Schmiedebergs Arch Pharmacol 358:145-152, 1998 12. Bartsch W: Anabolic steroids--action on cellular level. Wien Med Wochenschr 143:363-366, 1993 13. Basaria S, Wahlstrom JT, Dobs AS: Clinical review 138: Anabolic-androgenic steroid therapy in the treatment of chronic diseases. J Clin Endocrinol Metab 86:5108-5117, 2001 14. Bath CP, Farrell LN, Gilmore J, et al: The effects of ifenprodil and on voltage-dependent Ca2+ channels and in gerbil global cerebral ischaemia. Eur J Pharmacol 299:103-112, 1996 15. Baulieu EE: Neuroactive neurosteroids: dehydroepiandrosterone (DHEA) and DHEA sulphate. Acta Paediatr Suppl 88:78-80, 1999

58 16. Baulieu EE: Neurosteroids: a novel function of the brain. Psychoneuroendo- crinology 23:963-987, 1998 17. Baulieu EE, Robel P, Fellous A, et al: MAPREG: toward a novel approach of neuroprotection and treatment of Alzheimer's disease. J Mol Neurosci 24:63- 65, 2004 18. Beart PM, Mercer LD, Jarrott B: [125I]Ifenprodil: a convenient radioligand for binding and autoradiographic studies of the polyamine-sensitive site of the NMDA receptor. Neurosci Lett 124:187-189, 1991 19. Belelli D, Lambert JJ: Neurosteroids: endogenous regulators of the GABA(A) receptor. Nat Rev Neurosci 6:565-575, 2005 20. Bergeron R, de Montigny C, Debonnel G: Potentiation of neuronal NMDA response induced by dehydroepiandrosterone and its suppression by progester- one: effects mediated via sigma receptors. J Neurosci 16:1193-1202, 1996 21. Bergeron R, de Montigny C, Debonnel G: Pregnancy reduces brain sigma receptor function. Br J Pharmacol 127:1769-1776, 1999 22. Bergeron R, Debonnel G, De Montigny C: Modification of the N-methyl-D- aspartate response by sigma receptor ligands. Eur J Pharmacol 240:319-323, 1993 23. Bernardi F, Lanzone A, Cento RM, et al: Allopregnanolone and dehydroepian- drosterone response to corticotropin-releasing factor in patients suffering from Alzheimer's disease and vascular dementia. Eur J Endocrinol 142:466-471, 2000 24. Bhasin S, Storer TW, Berman N, et al: The effects of supraphysiologic doses of testosterone on muscle size and strength in normal men. N Engl J Med 335:1- 7., 1996 25. Bickelman C, Ferries L, Eaton RP: Impotence related to use in a body builder. Response to clomiphene citrate. West J Med 162:158-160., 1995 26. Billiards SS, Nguyen PN, Scheerlinck JP, et al: Hypoxia potentiates endotoxin- induced allopregnanolone concentrations in the newborn brain. Biol Neonate 90:258-267, 2006 27. Birgner C, Kindlundh-Hogberg AM, Nyberg F, et al: Altered extracellular levels of DOPAC and HVA in the rat nucleus accumbens shell in response to sub- chronic nandrolone administration and a subsequent amphetamine challenge. Neurosci Lett 412:168-172, 2007 28. Bisaga A, Popik P: In search of a new pharmacological treatment for drug and alcohol addiction: N-methyl-D-aspartate (NMDA) antagonists. Drug Alcohol Depend 59:1-15, 2000 29. Bitran D, Kellogg CK, Hilvers RJ: Treatment with an anabolic-androgenic steroid affects anxiety-related behavior and alters the sensitivity of cortical GABAA receptors in the rat. Horm Behav 27:568-583, 1993 30. Bitran D, Shiekh M, McLeod M: Anxiolytic effect of progesterone is mediated by the neurosteroid allopregnanolone at brain GABAA receptors. J Neuroen- docrinol 7:171-177, 1995 31. Boada LD, Fernandez L, Zumbado M, et al: Identification of a specific binding site for the anabolic steroid stanozolol in male rat liver microsomes. J Pharma- col Exp Ther 279:1123-1129, 1996 32. Bonhaus DW, Yeh GC, Skaryak L, et al: Glycine regulation of the N-methyl-D- aspartate receptor-gated ion channel in hippocampal membranes. Mol Pharma- col 36:273-279, 1989 33. Borea PA, Varani K, Gessi S, et al: Binding thermodynamics at the human neuronal nicotine receptor. Biochem Pharmacol 55:1189-1197, 1998

59 34. Bouchard P, Quirion R: [3H]1,3-di(2-tolyl)guanidine and [3H](+) binding sites in the rat brain: autoradiographic visualization of the putative sigma1 and sigma2 receptor subtypes. Neuroscience 76:467-477, 1997 35. Breuer ME, McGinnis MY, Lumia AR, et al: Aggression in male rats receiving anabolic androgenic steroids: effects of social and environmental provocation. Horm Behav 40:409-418, 2001 36. Brewer GJ, Cotman CW: NMDA receptor regulation of neuronal morphology in cultured hippocampal neurons. Neurosci Lett 99:268-273, 1989 37. Cagnotto A, Bastone A, Mennini T: [3H](+)-pentazocine binding to rat brain sigma 1 receptors. Eur J Pharmacol 266:131-138, 1994 38. Callier S, Morissette M, Grandbois M, et al: Neuroprotective properties of 17beta-estradiol, progesterone, and raloxifene in MPTP C57Bl/6 mice. Synapse 41:131-138, 2001 39. Carlson MD, Penney JB, Jr., Young AB: NMDA, AMPA, and binding site changes in Alzheimer's disease visual cortex. Neurobiol Aging 14:343-352, 1993 40. Carron C, Jullien A, Bucher B: Synthesis and pharmacological properties of a series of 2-piperidino alkanol derivatives. Arzneimittelforschung 21:1992- 1998, 1971 41. Carter C, Rivy JP, Scatton B: Ifenprodil and SL 82.0715 are antagonists at the polyamine site of the N-methyl-D-aspartate (NMDA) receptor. Eur J Pharma- col 164:611-612, 1989 42. Ceccon M, Rumbaugh G, Vicini S: Distinct effect of pregnenolone sulfate on NMDA receptor subtypes. Neuropharmacology 40:491-500, 2001 43. Charalampopoulos I, Alexaki VI, Tsatsanis C, et al: Neurosteroids as endoge- nous inhibitors of neuronal cell apoptosis in aging. Ann N Y Acad Sci 1088:139-152, 2006 44. Charalampopoulos I, Tsatsanis C, Dermitzaki E, et al: Dehydroepiandrosterone and allopregnanolone protect sympathoadrenal medulla cells against apoptosis via antiapoptotic Bcl-2 proteins. Proc Natl Acad Sci U S A 101:8209-8214, 2004 45. Chenard BL, Bordner J, Butler TW, et al: (1S,2S)-1-(4-hydroxyphenyl)-2-(4- hydroxy-4-phenylpiperidino)-1-propanol: a potent new neuroprotectant which blocks N-methyl-D-aspartate responses. J Med Chem 38:3138-3145, 1995 46. Cheng Y, Prusoff WH: Relationship between the inhibition constant (K1) and the concentration of inhibitor which causes 50 per cent inhibition (I50) of an en- zymatic reaction. Biochem Pharmacol 22:3099-3108, 1973 47. Cheng ZX, Lan DM, Wu PY, et al: Neurosteroid dehydroepiandrosterone sulphate inhibits persistent sodium currents in rat medial prefrontal cortex via activation of sigma-1 receptors. Exp Neurol, 2007 48. Choi DW, Rothman SM: The role of glutamate neurotoxicity in hypoxic- ischemic neuronal death. Annu Rev Neurosci 13:171-182, 1990 49. Church J, Fletcher EJ, Baxter K, et al: Blockade by ifenprodil of high voltage- activated Ca2+ channels in rat and mouse cultured hippocampal pyramidal neu- rones: comparison with N-methyl-D-aspartate receptor antagonist actions. Br J Pharmacol 113:499-507, 1994 50. Clark AS, Henderson LP: Behavioral and physiological responses to anabolic- androgenic steroids. Neurosci Biobehav Rev 27:413-436, 2003 51. Coan EJ, Collingridge GL: Effects of , SKF 10,047 and related psychotomimetic agents on N-methyl-D-aspartate receptor mediated synaptic responses in rat hippocampal slices. Br J Pharmacol 91:547-556, 1987

60 52. Coan EJ, Collingridge GL: Magnesium ions block an N-methyl-D-aspartate receptor-mediated component of synaptic transmission in rat hippocampus. Neurosci Lett 53:21-26, 1985 53. Compagnone NA, Mellon SH: Neurosteroids: biosynthesis and function of these novel neuromodulators. Front Neuroendocrinol 21:1-56, 2000 54. Contreras PC, Bremer ME, Gray NM: Ifenprodil and SL 82.0715 potently inhibit binding of [3H](+)-3-PPP to sigma binding sites in rat brain. Neurosci Lett 116:190-193, 1990 55. Contreras PC, Bremer ME, Rao TS: GBR-12909 and fluspirilene potently inhibited binding of [3H] (+)3-PPP to sigma receptors in rat brain. Life Sci 47:PL133-137, 1990 56. Contreras PC, Quirion R, Gehlert DR, et al: Autoradiographic distribution of non-dopaminergic binding sites labeled by [3H]haloperidol in rat brain. Neuro- sci Lett 75:133-140, 1987 57. Cook P, James I: Cerebral vasodilators (second of two parts). N Engl J Med 305:1560-1564, 1981 58. Cooper EJ, Johnston GA, Edwards FA: Effects of a naturally occurring neuros- teroid on GABAA IPSCs during development in rat hippocampal or cerebellar slices. J Physiol 521 Pt 2:437-449, 1999 59. Coughenour LL, Barr BM: Use of trifluoroperazine isolates a [(3)H]Ifenprodil binding site in rat brain membranes with the pharmacology of the voltage- independent ifenprodil site on N-methyl-D-aspartate receptors containing NR2B subunits. J Pharmacol Exp Ther 296:150-159, 2001 60. Coughenour LL, Cordon JJ: Characterization of haloperidol and trifluperidol as subtype-selective N-methyl-D-aspartate (NMDA) receptor antagonists using [3H]TCP and [3H]ifenprodil binding in rat brain membranes. J Pharmacol Exp Ther 280:584-592, 1997 61. Cuppini R, Sartini S, Ambrogini P, et al: Control of neuron outgrowth by NMDA receptors. J Submicrosc Cytol Pathol 31:31-40, 1999 62. D'Astous M, Morissette M, Tanguay B, et al: Dehydroepiandrosterone (DHEA) such as 17beta-estradiol prevents MPTP-induced dopamine depletion in mice. Synapse 47:10-14, 2003 63. Daggett LP, Johnson EC, Varney MA, et al: The human N-methyl-D-aspartate receptor 2C subunit: genomic analysis, distribution in human brain, and func- tional expression. J Neurochem 71:1953-1968, 1998 64. Del Cerro S, Garcia-Estrada J, Garcia-Segura LM: Neuroactive steroids regulate astroglia morphology in hippocampal cultures from adult rats. Glia 14:65-71, 1995 65. DeMeyts P, Bainco AR, Roth J: Site-site interactions among insulin receptors. Characterization of the negative cooperativity. J Biol Chem 251:1877-1888, 1976 66. Dhir A, Kulkarni SK: Involvement of sigma-1 receptor modulation in the anti- depressant action of . Neurosci Lett 420:204-208, 2007 67. Dickerman RD, Schaller F, Prather I, et al: Sudden cardiac death in a 20-year- old bodybuilder using anabolic steroids. Cardiology 86:172-173, 1995 68. Dingledine R, Borges K, Bowie D, et al: The ion channels. Pharmacol Rev 51:7-61, 1999 69. Dingledine R, McBain CJ, McNamara JO: Excitatory amino acid receptors in epilepsy. Trends Pharmacol Sci 11:334-338, 1990 70. Dong Y, Fu YM, Sun JL, et al: Neurosteroid enhances glutamate release in rat prelimbic cortex via activation of alpha1-adrenergic and sigma1 receptors. Cell Mol Life Sci 62:1003-1014, 2005

61 71. Ebert B, Wong EH, Krogsgaard-Larsen P: Identification of a novel NMDA receptor in rat cerebellum. Eur J Pharmacol 208:49-52, 1991 72. Edinger KL, Frye CA: Testosterone's anti-anxiety and analgesic effects may be due in part to actions of its 5alpha-reduced metabolites in the hippocampus. Psychoneuroendocrinology 30:418-430, 2005 73. Eriksson M, Nilsson A, Samuelsson H, et al: On the role of NR3A in human NMDA receptors. Physiol Behav 92:54-59, 2007 74. Eriksson M, Samuelsson H, Samuelsson EB, et al: The NMDAR subunit NR3A interacts with microtubule-associated protein 1S in the brain. Biochem Biophys Res Commun 361:127-132, 2007 75. Fava M, Abraham M, Pava J, et al: Cardiovascular risk factors in depression. The role of anxiety and anger. Psychosomatics 37:31-37, 1996 76. Fava M, Rosenbaum JF, McCarthy M, et al: Anger attacks in depressed outpa- tients and their response to fluoxetine. Psychopharmacol Bull 27:275-279, 1991 77. Fava M, Rosenbaum JF, Pava JA, et al: Anger attacks in unipolar depression, Part 1: Clinical correlates and response to fluoxetine treatment. Am J Psychia- try 150:1158-1163, 1993 78. Ferrari W, Bertolini A, Mucci P, et al: [Action of anabolic steroids on the development of the genital system in the rabbit. VI. Anabolic and androgenic activity of 13 beta-ethyl-17 beta-hydroxy-17 alpha-ethyl-4-gonen-3-one (Norbo- letone) in the rat]. Boll Soc Ital Biol Sper 44:1310-1313, 1968 79. Flood JF, Morley JE, Roberts E: Memory-enhancing effects in male mice of pregnenolone and steroids metabolically derived from it. Proc Natl Acad Sci U S A 89:1567-1571, 1992 80. Flood JF, Morley JE, Roberts E: Pregnenolone sulfate enhances post-training memory processes when injected in very low doses into limbic system struc- tures: the amygdala is by far the most sensitive. Proc Natl Acad Sci U S A 92:10806-10810, 1995 81. Flood JF, Roberts E: Dehydroepiandrosterone sulfate improves memory in aging mice. Brain Res 448:178-181, 1988 82. Flood JF, Smith GE, Roberts E: Dehydroepiandrosterone and its sulfate enhance memory retention in mice. Brain Res 447:269-278, 1988 83. Frye CA, Reed TA: Androgenic neurosteroids: anti- effects in an animal model of epilepsy. Psychoneuroendocrinology 23:385-399, 1998 84. Frye CA, Rhodes ME: Estrogen-priming can enhance progesterone's anti- seizure effects in part by increasing hippocampal levels of allopregnanolone. Pharmacol Biochem Behav 81:907-916, 2005 85. Gagliardi RJ: Neuroprotection, excitotoxicity and NMDA antagonists. Arq Neuropsiquiatr 58:583-588, 2000 86. Gallagher MJ, Huang H, Lynch DR: Modulation of the N-methyl-D-aspartate receptor by haloperidol: NR2B-specific interactions. J Neurochem 70:2120- 2128, 1998 87. Gallagher MJ, Huang H, Pritchett DB, et al: Interactions between ifenprodil and the NR2B subunit of the N-methyl-D-aspartate receptor. J Biol Chem 271:9603-9611, 1996 88. Gardoni F, Di Luca M: New targets for pharmacological intervention in the synapse. Eur J Pharmacol 545:2-10, 2006 89. Geddes JW, Ulas J, Brunner LC, et al: Hippocampal excitatory amino acid receptors in elderly, normal individuals and those with Alzheimer's disease: non-N-methyl-D-aspartate receptors. Neuroscience 50:23-34, 1992

62 90. Georg A, Friedl A: Identification and characterization of two sigma-like binding sites in the mouse neuroblastoma x rat glioma hybrid cell line NG108-15. J Pharmacol Exp Ther 259:479-483, 1991 91. Gibbs TT, Farb DH: Dueling enigmas: neurosteroids and sigma receptors in the limelight. Sci STKE 2000:PE1, 2000 92. Gibson DA, Harris BR, Rogers DT, et al: Radioligand binding studies reveal is a more selective antagonist for a polyamine-site on the NMDA re- ceptor than arcaine or ifenprodil. Brain Res 952:71-77, 2002 93. Girdler SS, Straneva PA, Light KC, et al: Allopregnanolone levels and reactiv- ity to mental stress in premenstrual dysphoric disorder. Biol Psychiatry 49:788- 797, 2001 94. Gordon I, Weizman R, Rehavi M: [3H]GBR 12935 labels mainly the piperazine acceptor site in the rat prefrontal cortex. Brain Res 674:205-210, 1995 95. Gotti B, Duverger D, Bertin J, et al: Ifenprodil and SL 82.0715 as cerebral anti- ischemic agents. I. Evidence for efficacy in models of focal cerebral ischemia. J Pharmacol Exp Ther 247:1211-1221, 1988 96. Graham D, Darles G, Langer SZ: The neuroprotective properties of ifenprodil, a novel NMDA receptor antagonist, in neuronal cell culture toxicity studies. Eur J Pharmacol 226:373-376, 1992 97. Grandbois M, Morissette M, Callier S, et al: Ovarian steroids and raloxifene prevent MPTP-induced dopamine depletion in mice. Neuroreport 11:343-346, 2000 98. Greenamyre JT, Penney JB, Young AB, et al: Alterations in L-glutamate bind- ing in Alzheimer's and Huntington's diseases. Science 227:1496-1499, 1985 99. Griffin LD, Gong W, Verot L, et al: Niemann-Pick type C disease involves disrupted neurosteroidogenesis and responds to allopregnanolone. Nat Med 10:704-711, 2004 100. Grimwood S, Gilbert E, Ragan CI, et al: Modulation of 45Ca2+ influx into cells stably expressing recombinant human NMDA receptors by ligands acting at dis- tinct recognition sites. J Neurochem 66:2589-2595, 1996 101. Grimwood S, Richards P, Murray F, et al: Characterisation of N-methyl-D- aspartate receptor-specific [(3)H]Ifenprodil binding to recombinant human NR1a/NR2B receptors compared with native receptors in rodent brain mem- branes. J Neurochem 75:2455-2463, 2000 102. Guazzo EP, Kirkpatrick PJ, Goodyer IM, et al: Cortisol, dehydroepiandroster- one (DHEA), and DHEA sulfate in the cerebrospinal fluid of man: relation to blood levels and the effects of age. J Clin Endocrinol Metab 81:3951-3960, 1996 103. Gursoy E, Cardounel A, Kalimi M: Pregnenolone protects mouse hippocampal (HT-22) cells against glutamate and amyloid beta protein toxicity. Neurochem Res 26:15-21, 2001 104. Hagihara Y: [Action mechanism and effects of ifenprodil on the regional blood flow, especially on cerebrovascular circulation]. Nippon Yakurigaku Zasshi 71:709-725, 1975 105. Hanner M, Moebius FF, Flandorfer A, et al: Purification, molecular cloning, and expression of the mammalian sigma1-binding site. Proc Natl Acad Sci U S A 93:8072-8077, 1996 106. Harney SC, Frenguelli BG, Lambert JJ: Phosphorylation influences neurosteroid modulation of synaptic GABAA receptors in rat CA1 and dentate gyrus neu- rones. Neuropharmacology 45:873-883, 2003

63 107. Harrison NL, Simmonds MA: Quantitative studies on some antagonists of N- methyl D-aspartate in slices of rat cerebral cortex. Br J Pharmacol 84:381-391, 1985 108. Hashimoto K, London ED: Further characterization of [3H]ifenprodil binding to sigma receptors in rat brain. Eur J Pharmacol 236:159-163, 1993 109. Hashimoto K, London ED: Interactions of erythro-ifenprodil, threo-ifenprodil, erythro-iodoifenprodil, and eliprodil with subtypes of sigma receptors. Eur J Pharmacol 273:307-310, 1995 110. Hashimoto K, Mantione CR, Spada MR, et al: Further characterization of [3H]ifenprodil binding in rat brain. Eur J Pharmacol 266:67-77, 1994 111. Hashimoto K, Scheffel U, London ED: In vivo labeling of sigma receptors in mouse brain with [3H]4-phenyl-1-(4-phenylbutyl)piperidine. Synapse 20:85-90, 1995 112. Hayashi T, Maurice T, Su TP: Ca(2+) signaling via sigma(1)-receptors: novel regulatory mechanism affecting intracellular Ca(2+) concentration. J Pharma- col Exp Ther 293:788-798, 2000 113. Hayashi T, Su TP: Intracellular dynamics of sigma-1 receptors (sigma(1) bind- ing sites) in NG108-15 cells. J Pharmacol Exp Ther 306:726-733, 2003 114. Herkert M, Rottger S, Becker CM: The NMDA receptor subunit NR2B of neonatal rat brain: complex formation and enrichment in axonal growth cones. Eur J Neurosci 10:1553-1562, 1998 115. Herrling PL, Misbach-Lesenne B: Effects of clozapine in a selective muscarinic bioassay and on single cells of the rat hippocampus. Naunyn Schmiedebergs Arch Pharmacol 320:20-25, 1982 116. Herzog AG, Drislane FW, Schomer DL, et al: Differential effects of antiepilep- tic drugs on neuroactive steroids in men with epilepsy. Epilepsia 47:1945-1948, 2006 117. Higashi T, Sugitani H, Yagi T, et al: Studies on neurosteroids XVI. Levels of pregnenolone sulfate in rat brains determined by enzyme-linked immunosorbent assay not requiring solvolysis. Biol Pharm Bull 26:709-711, 2003 118. Hill AV: The Combinations of Haemoglobin with Oxygen and with . I. Biochem J 7:471-480, 1913 119. Hirai H, Kirsch J, Laube B, et al: The glycine binding site of the N-methyl-D- aspartate receptor subunit NR1: identification of novel determinants of co- agonist potentiation in the extracellular M3-M4 loop region. Proc Natl Acad Sci U S A 93:6031-6036, 1996 120. Hirst JJ, Palliser HK, Yates DM, et al: Neurosteroids in the fetus and neonate: Potential protective role in compromised . Neurochem Int, 2007 121. Ho HP, Olsson M, Westberg L, et al: The serotonin reuptake inhibitor fluoxetine reduces sex steroid-related aggression in female rats: an animal model of pre- menstrual ? Neuropsychopharmacology 24:502-510, 2001 122. Hofner G, Wanner KT: [3H]ifenprodil binding to NMDA receptors in porcine hippocampal brain membranes. Eur J Pharmacol 394:211-219, 2000 123. Horak M, Vlcek K, Chodounska H, et al: Subtype-dependence of N-methyl-d- aspartate receptor modulation by pregnenolone sulfate. Neuroscience, 2005 124. Horak M, Vlcek K, Chodounska H, et al: Subtype-dependence of N-methyl-D- aspartate receptor modulation by pregnenolone sulfate. Neuroscience 137:93- 102, 2006 125. Horak M, Vlcek K, Petrovic M, et al: Molecular mechanism of pregnenolone sulfate action at NR1/NR2B receptors. J Neurosci 24:10318-10325, 2004

64 126. Ilyin VI, Whittemore ER, Guastella J, et al: Subtype-selective inhibition of N- methyl-D-aspartate receptors by haloperidol. Mol Pharmacol 50:1541-1550, 1996 127. Irwin RP, Lin SZ, Rogawski MA, et al: Steroid potentiation and inhibition of N- methyl-D-aspartate receptor-mediated intracellular Ca++ responses: structure- activity studies. J Pharmacol Exp Ther 271:677-682, 1994 128. Irwin RP, Maragakis NJ, Rogawski MA, et al: Pregnenolone sulfate augments NMDA receptor mediated increases in intracellular Ca2+ in cultured rat hippo- campal neurons. Neurosci Lett 141:30-34, 1992 129. Itzhak Y: Different modulation of the binding to two phencyclidine (PCP) receptor subtypes: effects of N-methyl-D-aspartate agonists and antagonists. Neurosci Lett 104:314-319, 1989 130. Itzhak Y: Multiple affinity binding states of the sigma receptor: effect of GTP- binding protein-modifying agents. Mol Pharmacol 36:512-517, 1989 131. Jahr CE, Stevens CF: Glutamate activates multiple single channel conductances in hippocampal neurons. Nature 325:522-525, 1987 132. Jang MK, Mierke DF, Russek SJ, et al: A steroid modulatory domain on NR2B controls N-methyl-D-aspartate receptor proton sensitivity. Proc Natl Acad Sci U S A, 2004 133. Jansen KL, Faull RL, Dragunow M, et al: Alzheimer's disease: changes in hippocampal N-methyl-D-aspartate, quisqualate, neurotensin, , benzo- diazepine, serotonin and opioid receptors--an autoradiographic study. Neurosci- ence 39:613-627, 1990 134. Jellinck PH, Lee SJ, McEwen BS: Metabolism of dehydroepiandrosterone by rat hippocampal cells in culture: possible role of aromatization and 7-hydroxylation in neuroprotection. J Steroid Biochem Mol Biol 78:313-317, 2001 135. Jensen AA, Spalding TA: Allosteric modulation of G-protein coupled receptors. Eur J Pharm Sci 21:407-420, 2004 136. Johansson T, Le Greves P: The effect of dehydroepiandrosterone sulfate and allopregnanolone sulfate on the binding of [(3)H]ifenprodil to the N-methyl-d- aspartate receptor in rat frontal cortex membrane. J Steroid Biochem Mol Biol 94:263-266, 2005 137. Johnson JW, Ascher P: Glycine potentiates the NMDA response in cultured mouse brain neurons. Nature 325:529-531, 1987 138. Kaasik A, Kalda A, Jaako K, et al: Dehydroepiandrosterone sulphate prevents oxygen-glucose deprivation-induced injury in cerebellar granule cell culture. Neuroscience 102:427-432, 2001 139. Karasawa J, Yamamoto H, Yamamoto T, et al: MS-377, a selective sigma receptor ligand, indirectly blocks the action of PCP in the N-methyl-D-aspartate receptor ion-channel complex in primary cultured rat neuronal cells. Life Sci 70:1631-1642, 2002 140. Karbon EW, Patch RJ, Pontecorvo MJ, et al: Ifenprodil potently interacts with [3H](+)-3-PPP-labeled sigma binding sites in guinea pig brain membranes. Eur J Pharmacol 176:247-248, 1990 141. Kashiwagi H, McDunn JE, Simon PO, Jr., et al: Selective sigma-2 ligands preferentially bind to pancreatic adenocarcinomas: applications in diagnostic imaging and therapy. Mol Cancer 6:48, 2007 142. Kew JN, Kemp JA: An allosteric interaction between the NMDA receptor polyamine and ifenprodil sites in rat cultured cortical neurones. J Physiol 512 ( Pt 1):17-28, 1998

65 143. Kew JN, Richards JG, Mutel V, et al: Developmental changes in NMDA recep- tor glycine affinity and ifenprodil sensitivity reveal three distinct populations of NMDA receptors in individual rat cortical neurons. J Neurosci 18:1935-1943, 1998 144. Kew JN, Trube G, Kemp JA: A novel mechanism of activity-dependent NMDA receptor antagonism describes the effect of ifenprodil in rat cultured cortical neurones. J Physiol 497 ( Pt 3):761-772, 1996 145. Kilpatrick GJ, el Tayar N, Van de Waterbeemd H, et al: The thermodynamics of agonist and antagonist binding to dopamine D-2 receptors. Mol Pharmacol 30:226-234, 1986 146. Kilpatrick GJ, Jenner P, Marsden CD: Properties of rat striatal D-2 dopamine receptors solubilized with the zwitterionic detergent CHAPS. J Pharm Phar- macol 37:320-328, 1985 147. Kindlundh AM, Lindblom J, Bergstrom L, et al: The anabolic-androgenic steroid nandrolone induces alterations in the density of serotonergic 5HT1B and 5HT2 receptors in the male rat brain. Neuroscience 119:113-120, 2003 148. Kindlundh AM, Lindblom J, Bergstrom L, et al: The anabolic-androgenic steroid nandrolone decanoate affects the density of dopamine receptors in the male rat brain. Eur J Neurosci 13:291-296, 2001 149. Klotz F, Garle M, Granath F, et al: Criminality among individuals testing posi- tive for the presence of anabolic androgenic steroids. Arch Gen Psychiatry 63:1274-1279, 2006 150. Kresse W, Sekler I, Hoffmann A, et al: Zinc ions are endogenous modulators of -stimulated capacitative Ca2+ entry in both cultured and in situ mouse astrocytes. Eur J Neurosci 21:1626-1634, 2005 151. Kuryatov A, Laube B, Betz H, et al: Mutational analysis of the glycine-binding site of the NMDA receptor: structural similarity with bacterial amino acid- binding proteins. Neuron 12:1291-1300, 1994 152. Kutsuwada T, Sakimura K, Manabe T, et al: Impairment of suckling response, trigeminal neuronal pattern formation, and hippocampal LTD in NMDA recep- tor epsilon 2 subunit mutant mice. Neuron 16:333-344, 1996 153. Lambert JJ, Belelli D, Harney SC, et al: Modulation of native and recombinant GABA(A) receptors by endogenous and synthetic neuroactive steroids. Brain Res Brain Res Rev 37:68-80, 2001 154. Lapchak PA, Chapman DF, Nunez SY, et al: Dehydroepiandrosterone sulfate is neuroprotective in a reversible spinal cord ischemia model: possible involve- ment of GABA(A) receptors. Stroke 31:1953-1956; discussion 1957, 2000 155. Lau LF, Huganir RL: Differential tyrosine phosphorylation of N-methyl-D- aspartate receptor subunits. J Biol Chem 270:20036-20041, 1995 156. Laube B, Hirai H, Sturgess M, et al: Molecular determinants of agonist dis- crimination by NMDA receptor subunits: analysis of the glutamate binding site on the NR2B subunit. Neuron 18:493-503, 1997 157. Le Greves P, Huang W, Johansson P, et al: 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 226:61-64, 1997 158. Le Greves P, Zhou Q, Huang W, et al: Effect of combined treatment with nandrolone and cocaine on the NMDA receptor gene expression in the rat nu- cleus accumbens and periaqueductal gray. Acta Psychiatr Scand Suppl:129- 132, 2002 159. Legendre P, Westbrook GL: Ifenprodil blocks N-methyl-D-aspartate receptors by a two-component mechanism. Mol Pharmacol 40:289-298, 1991

66 160. Legrain S, Berr C, Frenoy N, et al: Dehydroepiandrosterone sulfate in a long- term care aged population. Gerontology 41:343-351, 1995 161. Legrain S, Girard L: Pharmacology and therapeutic effects of dehydroepian- drosterone in older subjects. Drugs Aging 20:949-967, 2003 162. Leysen JE, Gommeren W: Optimal conditions for [3H]apomorphine binding and anomalous equilibrium binding of [3H]apomorphine and [3H]spiperone to rat striatal membranes: involvement of surface phenomena versus multiple bind- ing sites. J Neurochem 36:201-219, 1981 163. Limbird L: Cell surface receptors A short course on theory and methods, ed 3. New York: Springer, 2004 164. Lindstrom M, Nilsson AL, Katzman PL, et al: Use of anabolic-androgenic steroids among body builders--frequency and attitudes. J Intern Med 227:407- 411, 1990 165. Liu S, Sjovall J, Griffiths WJ: Neurosteroids in rat brain: extraction, isolation, and analysis by nanoscale liquid chromatography-electrospray mass spectrome- try. Anal Chem 75:5835-5846, 2003 166. Liu Y, Chen GD, Lerner MR, et al: Cocaine up-regulates Fra-2 and sigma-1 receptor gene and protein expression in brain regions involved in addiction and reward. J Pharmacol Exp Ther 314:770-779, 2005 167. Low CM, Zheng F, Lyuboslavsky P, et al: Molecular determinants of coordi- nated proton and zinc inhibition of N-methyl-D-aspartate NR1/NR2A receptors. Proc Natl Acad Sci U S A 97:11062-11067, 2000 168. Lowry OH, Rosebrough NJ, Farr AL, et al: Protein measurement with the Folin phenol reagent. J Biol Chem 193:265-275, 1951 169. Lynch G: Long-term potentiation in the Eocene. Philos Trans R Soc Lond B Biol Sci 358:625-628, 2003 170. Maeda Y, Furukawa T: [Phamacolofical studies of ifenprodil]. Nippon Yakuri- gaku Zasshi 71:585-595, 1975 171. Majewska MD: Neurosteroids: endogenous bimodal modulators of the GABAA receptor. Mechanism of action and physiological significance. Prog Neurobiol 38:379-395, 1992 172. Majewska MD, Bisserbe JC, Eskay RL: Glucocorticoids are modulators of GABAA receptors in brain. Brain Res 339:178-182, 1985 173. Maksay G: Thermodynamics of gamma-aminobutyric acid type A receptor binding differentiate agonists from antagonists. Mol Pharmacol 46:386-390, 1994 174. Malayev A, Gibbs TT, Farb DH: Inhibition of the NMDA response by pregne- nolone sulphate reveals subtype selective modulation of NMDA receptors by sulphated steroids. Br J Pharmacol 135:901-909, 2002 175. Mameli M, Carta M, Partridge LD, et al: Neurosteroid-induced plasticity of immature synapses via retrograde modulation of presynaptic NMDA receptors. J Neurosci 25:2285-2294, 2005 176. Martin WR, Eades CG, Thompson JA, et al: The effects of - and nalorphine- like drugs in the nondependent and morphine-dependent chronic spinal dog. J Pharmacol Exp Ther 197:517-532, 1976 177. Martina M, Turcotte ME, Halman S, et al: The sigma-1 receptor modulates NMDA receptor synaptic transmission and plasticity via SK channels in rat hip- pocampus. J Physiol 578:143-157, 2007 178. Marx CE, Trost WT, Shampine LJ, et al: The neurosteroid allopregnanolone is reduced in prefrontal cortex in Alzheimer's disease. Biol Psychiatry 60:1287- 1294, 2006

67 179. Masuko T, Kashiwagi K, Kuno T, et al: A regulatory domain (R1-R2) in the amino terminus of the N-methyl-D-aspartate receptor: effects of , pro- tons, and ifenprodil, and structural similarity to bacterial leu- cine/isoleucine/valine binding protein. Mol Pharmacol 55:957-969, 1999 180. Mathis C, Paul SM, Crawley JN: The neurosteroid pregnenolone sulfate blocks NMDA antagonist-induced deficits in a passive avoidance memory task. Psy- chopharmacology (Berl) 116:201-206, 1994 181. Mathis C, Vogel E, Cagniard B, et al: The neurosteroid pregnenolone sulfate blocks deficits induced by a competitive NMDA antagonist in active avoidance and lever-press learning tasks in mice. Neuropharmacology 35:1057-1064, 1996 182. Matsuno K, Kobayashi T, Tanaka MK, et al: Sigma 1 receptor subtype is in- volved in the relief of behavioral despair in the mouse forced swimming test. Eur J Pharmacol 312:267-271, 1996 183. Maurice T, Hiramatsu M, Itoh J, et al: Behavioral evidence for a modulating role of sigma ligands in memory processes. I. Attenuation of dizocilpine (MK- 801)-induced amnesia. Brain Res 647:44-56, 1994 184. Maurice T, Hiramatsu M, Itoh J, et al: Low dose of 1,3-di(2-tolyl)guanidine (DTG) attenuates MK-801-induced spatial working memory impairment in mice. Psychopharmacology (Berl) 114:520-522, 1994 185. Maurice T, Hiramatsu M, Kameyama T, et al: Behavioral evidence for a modu- lating role of sigma ligands in memory processes. II. Reversion of carbon mon- oxide-induced amnesia. Brain Res 647:57-64, 1994 186. Maurice T, Junien JL, Privat A: Dehydroepiandrosterone sulfate attenuates dizocilpine-induced learning impairment in mice via sigma 1-receptors. Behav Brain Res 83:159-164, 1997 187. Maurice T, Lockhart BP, Su TP, et al: Reversion of beta 25-35-amyloid peptide- induced amnesia by NMDA receptor-associated glycine site agonists. Brain Res 731:249-253, 1996 188. Maurice T, Phan VL, Urani A, et al: Differential involvement of the sigma(1) (sigma(1)) receptor in the anti-amnesic effect of neuroactive steroids, as demon- strated using an in vivo antisense strategy in the mouse. Br J Pharmacol 134:1731-1741, 2001 189. Maurice T, Phan VL, Urani A, et al: Neuroactive neurosteroids as endogenous effectors for the sigma1 (sigma1) receptor: pharmacological evidence and thera- peutic opportunities. Jpn J Pharmacol 81:125-155, 1999 190. Maurice T, Su TP, Parish DW, et al: PRE-084, a sigma selective PCP deriva- tive, attenuates MK-801-induced impairment of learning in mice. Pharmacol Biochem Behav 49:859-869, 1994 191. Maurice T, Urani A, Phan VL, et al: The interaction between neuroactive ster- oids and the sigma1 receptor function: behavioral consequences and therapeutic opportunities. Brain Res Brain Res Rev 37:116-132, 2001 192. Mayer ML, Westbrook GL, Guthrie PB: Voltage-dependent block by Mg2+ of NMDA responses in spinal cord neurones. Nature 309:261-263, 1984 193. Mayo W, Dellu F, Robel P, et al: Infusion of neurosteroids into the nucleus basalis magnocellularis affects cognitive processes in the rat. Brain Res 607:324-328, 1993 194. McCool BA, Lovinger DM: Ifenprodil inhibition of the 5-hydroxytryptamine3 receptor. Neuropharmacology 34:621-629, 1995

68 195. McLaren S, Cookson JC, Silverstone T: Positive and negative symptoms, depression and social disability in chronic schizophrenia: a comparative trial of bromperidol and decanoates. Int Clin Psychopharmacol 7:67-72, 1992 196. McLennan H, Hicks TP, Liu JR: On the configuration of the receptors for excitatory amino acids. Neuropharmacology 21:549-554, 1982 197. Mellon SH, Griffin LD, Compagnone NA: Biosynthesis and action of neuros- teroids. Brain Res Brain Res Rev 37:3-12, 2001 198. Menegoz M, Lau LF, Herve D, et al: Tyrosine phosphorylation of NMDA receptor in rat striatum: effects of 6-OH-dopamine lesions. Neuroreport 7:125- 128, 1995 199. Mensah-Nyagan AG, Do-Rego JL, Beaujean D, et al: Neurosteroids: expression of steroidogenic enzymes and regulation of steroid biosynthesis in the central nervous system. Pharmacol Rev 51:63-81, 1999 200. Meyer DA, Carta M, Partridge LD, et al: Neurosteroids enhance spontaneous glutamate release in hippocampal neurons. Possible role of metabotropic sigma1-like receptors. J Biol Chem 277:28725-28732, 2002 201. Meziane H, Mathis C, Paul SM, et al: The neurosteroid pregnenolone sulfate reduces learning deficits induced by scopolamine and has promnestic effects in mice performing an appetitive learning task. Psychopharmacology (Berl) 126:323-330, 1996 202. Monaghan DT, Holets VR, Toy DW, et al: Anatomical distributions of four pharmacologically distinct 3H-L-glutamate binding sites. Nature 306:176-179, 1983 203. Monnet FP, Blier P, Debonnel G, et al: Modulation by sigma ligands of N- methyl-D-aspartate-induced [3H]noradrenaline release in the rat hippocampus: G-protein dependency. Naunyn Schmiedebergs Arch Pharmacol 346:32-39, 1992 204. Monnet FP, Mahe V, Robel P, et al: Neurosteroids, via sigma receptors, modu- late the [3H] release evoked by N-methyl-D-aspartate in the rat hippocampus. Proc Natl Acad Sci U S A 92:3774-3778, 1995 205. Monnet FP, Maurice T: The sigma1 protein as a target for the non-genomic effects of neuro(active)steroids: molecular, physiological, and behavioral as- pects. J Pharmacol Sci 100:93-118, 2006 206. Monyer H, Burnashev N, Laurie DJ, et al: Developmental and regional expres- sion in the rat brain and functional properties of four NMDA receptors. Neuron 12:529-540, 1994 207. Monyer H, Sprengel R, Schoepfer R, et al: Heteromeric NMDA receptors: molecular and functional distinction of subtypes. Science 256:1217-1221, 1992 208. Morales AJ, Haubrich RH, Hwang JY, et al: The effect of six months treatment with a 100 mg daily dose of dehydroepiandrosterone (DHEA) on circulating sex steroids, body composition and muscle strength in age-advanced men and women. Clin Endocrinol (Oxf) 49:421-432, 1998 209. Morfin R, Starka L: Neurosteroid 7-hydroxylation products in the brain. Int Rev Neurobiol 46:79-95, 2001 210. Mott DD, Doherty JJ, Zhang S, et al: Phenylethanolamines inhibit NMDA receptors by enhancing proton inhibition. Nat Neurosci 1:659-667, 1998 211. Mukai H, Uchino S, Kawato S: Effects of neurosteroids on Ca(2+) signaling mediated by recombinant N-methyl-D-aspartate receptor expressed in Chinese hamster ovary cells. Neurosci Lett 282:93-96, 2000

69 212. Mukhin A, Kovaleva ES, London ED: Two affinity states of N-methyl-D- aspartate recognition sites: modulation by cations. J Pharmacol Exp Ther 282:945-954, 1997 213. Nacher J, McEwen BS: The role of N-methyl-D-asparate receptors in neuro- genesis. Hippocampus 16:267-270, 2006 214. Nacher J, Varea E, Miguel Blasco-Ibanez J, et al: N-methyl-d-aspartate receptor expression during in the rat dentate gyrus. Neuroscience 144:855-864, 2007 215. Narita M, Soma M, Mizoguchi H, et al: Implications of the NR2B subunit- containing NMDA receptor localized in mouse limbic forebrain in ethanol de- pendence. Eur J Pharmacol 401:191-195, 2000 216. Nateri AS, Raivich G, Gebhardt C, et al: ERK activation causes epilepsy by stimulating NMDA receptor activity. Embo J 26:4891-4901, 2007 217. Nicolas C, Carter C: Autoradiographic distribution and characteristics of high- and low-affinity polyamine-sensitive [3H]ifenprodil sites in the rat brain: possi- ble relationship to NMDAR2B receptors and calmodulin. J Neurochem 63:2248-2258, 1994 218. Nilsen J, Brinton RD: Impact of progestins on estrogen-induced neuroprotec- tion: synergy by progesterone and 19-norprogesterone and antagonism by me- droxyprogesterone acetate. Endocrinology 143:205-212, 2002 219. Nilsson A, Eriksson M, Muly EC, et al: Analysis of NR3A receptor subunits in human native NMDA receptors. Brain Res, 2007 220. Nixon RA: Niemann-Pick Type C disease and Alzheimer's disease: the APP- endosome connection fattens up. Am J Pathol 164:757-761, 2004 221. Noda Y, Nabeshima T: Opiate physical dependence and N-methyl-D-aspartate receptors. Eur J Pharmacol 500:121-128, 2004 222. Nowak L, Bregestovski P, Ascher P, et al: Magnesium gates glutamate-activated channels in mouse central neurones. Nature 307:462-465, 1984 223. Nyberg S, Andersson A, Zingmark E, et al: The effect of a low dose of alcohol on allopregnanolone serum concentrations across the in women with severe and controls. Psychoneuroendocrinology 30:892-901, 2005 224. Obara M, Szeliga M, Albrecht J: Regulation of pH in the mammalian central nervous system under normal and pathological conditions: Facts and hypothe- ses. Neurochem Int, 2007 225. Ohkuma S, Katsura M, Chen DZ, et al: Presence of N-methyl-D-aspartate (NMDA) receptors in neuroblastoma x glioma hybrid NG108-15 cells-analysis using [45Ca2+]influx and [3H]MK-801 binding as functional measures. Brain Res Mol Brain Res 22:166-172, 1994 226. Olney JW, Farber NB: Glutamate receptor dysfunction and schizophrenia. Arch Gen Psychiatry 52:998-1007, 1995 227. Olney JW, Wozniak DF, Farber NB: Glumate receptor dysfunction and Alz- heimer's disease. Restor Neurol Neurosci 13:75-83, 1998 228. Oswald RE: Ionotropic glutamate receptor recognition and activation, in Garcia KC (ed): Cell surface receptors. San Diego: Elsevier Aca, 2004, Vol 68, pp 314-341 229. Pahk AJ, Williams K: Influence of extracellular pH on inhibition by ifenprodil at N-methyl-D-aspartate receptors in Xenopus oocytes. Neurosci Lett 225:29- 32, 1997 230. Palmer AM, Burns MA: Preservation of redox, polyamine, and glycine modula- tory domains of the N-methyl-D-aspartate receptor in Alzheimer's disease. J Neurochem 62:187-196, 1994

70 231. Park-Chung M, Wu FS, Farb DH: 3 alpha-Hydroxy-5 beta-pregnan-20-one sulfate: a negative modulator of the NMDA-induced current in cultured neurons. Mol Pharmacol 46:146-150, 1994 232. Park-Chung M, Wu FS, Purdy RH, et al: Distinct sites for inverse modulation of N-methyl-D-aspartate receptors by sulfated steroids. Mol Pharmacol 52:1113- 1123, 1997 233. Pasternak GW, Kolesnikov YA, Babey AM: Perspectives on the N-methyl-D- aspartate/ cascade and opioid tolerance. Neuropsychopharmacol- ogy 13:309-313, 1995 234. Pearce IA, Cambray-Deakin MA, Burgoyne RD: Glutamate acting on NMDA receptors stimulates neurite outgrowth from cerebellar granule cells. FEBS Lett 223:143-147, 1987 235. Pearson H: Hormone therapy: a dangerous elixir? Nature 431:500-501, 2004 236. Petersson A, Garle M, Granath F, et al: Morbidity and mortality in patients testing positively for the presence of anabolic androgenic steroids in connection with receiving medical care. A controlled retrospective cohort study. Drug Al- cohol Depend 81:215-220, 2006 237. Petralia SM, DeBold JF, Frye CA: MK-801 infusions to the ventral tegmental area and ventromedial hypothalamus produce opposite effects on lordosis of hormone-primed rats. Pharmacol Biochem Behav 86:377-385, 2007 238. Phan VL, Miyamoto Y, Nabeshima T, et al: Age-related expression of sigma1 receptors and antidepressant efficacy of a selective agonist in the senescence- accelerated (SAM) mouse. J Neurosci Res 79:561-572, 2005 239. Pibiri F, Nelson M, Carboni G, et al: Neurosteroids regulate mouse aggression induced by anabolic androgenic steroids. Neuroreport 17:1537-1541, 2006 240. Pickles VR: Neurosteroids and aggressive behaviour in Alzheimer's disease. Med Hypotheses 42:243-244, 1994 241. Pinna G, Agis-Balboa RC, Doueiri MS, et al: Brain neurosteroids in gender- related aggression induced by social isolation. Crit Rev Neurobiol 16:75-82, 2004 242. Pinna G, Costa E, Guidotti A: Changes in brain testosterone and allopreg- nanolone biosynthesis elicit aggressive behavior. Proc Natl Acad Sci U S A 102:2135-2140, 2005 243. Pinna G, Costa E, Guidotti A: Fluoxetine and norfluoxetine stereospecifically and selectively increase brain neurosteroid content at doses that are inactive on 5-HT reuptake. Psychopharmacology (Berl) 186:362-372, 2006 244. Pinna G, Dong E, Matsumoto K, et al: In socially isolated mice, the reversal of brain allopregnanolone down-regulation mediates the anti-aggressive action of fluoxetine. Proc Natl Acad Sci U S A 100:2035-2040, 2003 245. Pontecorvo MJ, Karbon EW, Goode S, et al: Possible cerebroprotective and in vivo NMDA antagonist activities of sigma agents. Brain Res Bull 26:461-465, 1991 246. Pope HG, Jr., Katz DL: Psychiatric and medical effects of anabolic-androgenic steroid use. A controlled study of 160 athletes. Arch Gen Psychiatry 51:375- 382, 1994 247. Pringle AK, Schmidt W, Deans JK, et al: 7-Hydroxylated epiandrosterone (7- OH-EPIA) reduces ischaemia-induced neuronal damage both in vivo and in vi- tro. Eur J Neurosci 18:117-124, 2003 248. Quirion R, Bowen WD, Itzhak Y, et al: A proposal for the classification of sigma binding sites. Trends Pharmacol Sci 13:85-86, 1992 249. Rachline J, Perin-Dureau F, Le Goff A, et al: The micromolar zinc-binding domain on the NMDA receptor subunit NR2B. J Neurosci 25:308-317, 2005

71 250. Raikkonen K, Matthews KA, Salomon K: Hostility predicts metabolic syndrome risk factors in children and adolescents. Health Psychol 22:279-286, 2003 251. Ramachandran S, Lu H, Prabhu U, et al: Purification and characterization of the guinea pig sigma-1 receptor functionally expressed in Escherichia coli. Protein Expr Purif 51:283-292, 2007 252. Rang HP, Dale, M.M., Ritter, J.M.: Pharmacology, ed 3rd. New York: Chur- chill Livingstone, 1995 253. Ransom RW: Polyamine and ifenprodil interactions with the NMDA receptor's glycine site. Eur J Pharmacol 208:67-71, 1991 254. Rashid NA, Cambray-Deakin MA: N-methyl-D-aspartate effects on the growth, morphology and cytoskeleton of individual neurons in vitro. Brain Res Dev Brain Res 67:301-308, 1992 255. Reddy DS, Kaur G, Kulkarni SK: Sigma (sigma1) receptor mediated anti- depressant-like effects of neurosteroids in the Porsolt forced swim test. Neu- roreport 9:3069-3073, 1998 256. Reynolds IJ, Miller RJ: Ifenprodil is a novel type of N-methyl-D-aspartate receptor antagonist: interaction with polyamines. Mol Pharmacol 36:758-765, 1989 257. Rivera-Arce JC, Morales-Crespo L, Vargas-Pinto N, et al: Central effects of the anabolic steroid 17alpha methyltestosterone in female anxiety. Pharmacol Bio- chem Behav 84:275-281, 2006 258. Rocha VM, Calil CM, Ferreira R, et al: Influence of anabolic steroid on anxiety levels in sedentary male rats. Stress:1, 2007 259. Rock DM, Macdonald RL: Polyamine regulation of N-methyl-D-aspartate receptor channels. Annu Rev Pharmacol Toxicol 35:463-482, 1995 260. Romeo E, Strohle A, Spalletta G, et al: Effects of antidepressant treatment on neuroactive steroids in major depression. Am J Psychiatry 155:910-913, 1998 261. Roose K: as a replacement for maintenance therapy with intramuscular fluphenazine decanoate in schizophrenia and other chronic psy- choses. Acta Psychiatr Belg 82:216-223, 1982 262. Sanchez C, Arnt J, Costall B, et al: The selective sigma2-ligand Lu 28-179 has potent anxiolytic-like effects in rodents. J Pharmacol Exp Ther 283:1323- 1332, 1997 263. Sanchez C, Meier E: Behavioral profiles of SSRIs in animal models of depres- sion, anxiety and aggression. Are they all alike? Psychopharmacology (Berl) 129:197-205, 1997 264. Santner SJ, Santen RJ: Inhibition of estrone sulfatase and 17 beta- hydroxysteroid dehydrogenase by antiestrogens. J Steroid Biochem Mol Biol 45:383-390, 1993 265. Scatton B, Avenet P, Benavides J, et al: Neuroprotective potential of the poly- amine site-directed NMDA receptor antagonists ifenprodil and eliprodil., in Pal- freyman MG, Reynolds IJ, Skolnick P (eds): Direct and Allosteric Control of Glutamate Receptors: CRC Press, 1994, pp 139-154 266. Schaeffer V, Patte-Mensah C, Eckert A, et al: Modulation of neurosteroid production in human neuroblastoma cells by Alzheimer's disease key proteins. J Neurobiol 66:868-881, 2006 267. Schiess AR, Partridge LD: Pregnenolone sulfate acts through a G-protein- coupled sigma1-like receptor to enhance short term facilitation in adult hippo- campal neurons. Eur J Pharmacol 518:22-29, 2005 268. Schoemaker H, Allen J, Langer SZ: Binding of [3H]ifenprodil, a novel NMDA antagonist, to a polyamine-sensitive site in the rat cerebral cortex. Eur J Phar- macol 176:249-250, 1990

72 269. Schumacher M, Liere P, Akwa Y, et al: Pregnenolone sulfate in the brain: A controversial neurosteroid. Neurochem Int, 2007 270. Seidman KJ, Barsuk JH, Johnson RF, et al: Differentiation of NG108-15 neuro- blastoma cells by serum starvation or dimethyl sulfoxide results in marked dif- ferences in angiotensin II receptor subtype expression. J Neurochem 66:1011- 1018, 1996 271. Seth P, Fei YJ, Li HW, et al: Cloning and functional characterization of a sigma receptor from rat brain. J Neurochem 70:922-931, 1998 272. Shalaby IA, Chenard BL, Prochniak MA, et al: Neuroprotective effects of the N-methyl-D-aspartate receptor antagonists ifenprodil and SL-82,0715 on hippo- campal cells in culture. J Pharmacol Exp Ther 260:925-932, 1992 273. Shaw GG, Pateman AJ: The regional distribution of the polyamines spermidine and spermine in brain. J Neurochem 20:1225-1230, 1973 274. Sheng M, Cummings J, Roldan LA, et al: Changing subunit composition of heteromeric NMDA receptors during development of rat cortex. Nature 368:144-147, 1994 275. Shimazu S, Katsuki H, Takenaka C, et al: sigma receptor ligands attenuate N- methyl-D-aspartate cytotoxicity in dopaminergic neurons of mesencephalic slice cultures. Eur J Pharmacol 388:139-146, 2000 276. Shimizu E, Tang YP, Rampon C, et al: NMDA receptor-dependent synaptic reinforcement as a crucial process for memory consolidation. Science 290:1170-1174, 2000 277. Skuza G, Rogoz Z: A potential antidepressant activity of SA4503, a selective sigma 1 receptor agonist. Behav Pharmacol 13:537-543, 2002 278. Skuza G, Rogoz Z: The synergistic effect of selective sigma receptor agonists and uncompetitive NMDA receptor antagonists in the forced swim test in rats. J Physiol Pharmacol 57:217-229, 2006 279. Smith CD, Wekstein DR, Markesbery WR, et al: 3alpha,5alpha-THP: a poten- tial plasma neurosteroid biomarker in Alzheimer's disease and perhaps non- Alzheimer's dementia. Psychopharmacology (Berl) 186:481-485, 2006 280. Snyder PJ: , in Hardman JG, Limbird, L. E. (ed): The Pharmacol- ogical basis of therapeutics, ed 10th. New York: McGraw-Hill, 2001, pp 1635-1648 281. Spivak V, Lin A, Beebe P, et al: Identification of a neurosteroid binding site contained within the GluR2-S1S2 domain. Lipids 39:811-819, 2004 282. Steele JE, Palmer AM, Stratmann GC, et al: The N-methyl-D-aspartate receptor complex in Alzheimer's disease: reduced regulation by glycine but not zinc. Brain Res 500:369-373, 1989 283. Stieg PE, Sathi S, Warach S, et al: Neuroprotection by the NMDA receptor- associated open-channel blocker memantine in a photothrombotic model of cerebral focal ischemia in neonatal rat. Eur J Pharmacol 375:115-120, 1999 284. Su TP: Evidence for sigma opioid receptor: binding of [3H]SKF-10047 to etorphine-inaccessible sites in guinea-pig brain. J Pharmacol Exp Ther 223:284-290, 1982 285. Sullivan JM, Traynelis SF, Chen HS, et al: Identification of two cysteine resi- dues that are required for redox modulation of the NMDA subtype of glutamate receptor. Neuron 13:929-936, 1994 286. Takebayashi M, Hayashi T, Su TP: Sigma-1 receptors potentiate epidermal growth factor signaling towards neuritogenesis in PC12 cells: potential relation to lipid raft reconstitution. Synapse 53:90-103, 2004

73 287. Tam SW, Cook L: Sigma opiates and certain antipsychotic drugs mutually inhibit (+)-[3H] SKF 10,047 and [3H]haloperidol binding in guinea pig brain membranes. Proc Natl Acad Sci U S A 81:5618-5621, 1984 288. Tamura Y, Sato Y, Yokota T, et al: Ifenprodil prevents glutamate cytotoxicity via polyamine modulatory sites of N-methyl-D-aspartate receptors in cultured cortical neurons. J Pharmacol Exp Ther 265:1017-1025, 1993 289. Tang CM, Dichter M, Morad M: Modulation of the N-methyl-D-aspartate channel by extracellular H+. Proc Natl Acad Sci U S A 87:6445-6449, 1990 290. Thiblin I, Lindquist O, Rajs J: Cause and manner of death among users of anabolic androgenic steroids. J Forensic Sci 45:16-23, 2000 291. Thiblin I, Parlklo T: Anabolic androgenic steroids and violence. Acta Psychiatr Scand Suppl:125-128, 2002 292. Thiblin I, Petersson A: Pharmacoepidemiology of anabolic androgenic steroids: a review. Fundam Clin Pharmacol 19:27-44, 2005 293. Thiblin I, Runeson B, Rajs J: Anabolic androgenic steroids and suicide. Ann Clin Psychiatry 11:223-231, 1999 294. Thomson AM, Lodge D: Selective blockade of an excitatory synapse in rat cerebral cortex by the sigma opiate : an intracellular, in vitro study. Neurosci Lett 54:21-26, 1985 295. Tokuyama S, Hirata K, Yoshida A, et al: Selective coupling of mouse brain metabotropic sigma receptor with recombinant Gi1. Neurosci Lett 268:85-88, 1999 296. Tovar KR, Sprouffske K, Westbrook GL: Fast NMDA receptor-mediated synaptic currents in neurons from mice lacking the epsilon2 (NR2B) subunit. J Neurophysiol 83:616-620, 2000 297. Traynelis SF, Cull-Candy SG: Proton inhibition of N-methyl-D-aspartate recep- tors in cerebellar neurons. Nature 345:347-350, 1990 298. Traynelis SF, Hartley M, Heinemann SF: Control of proton sensitivity of the NMDA receptor by RNA splicing and polyamines. Science 268:873-876, 1995 299. Trujillo KA: Are NMDA receptors involved in opiate-induced neural and behavioral plasticity? A review of preclinical studies. Psychopharmacology (Berl) 151:121-141, 2000 300. Trujillo KA: Effects of noncompetitive N-methyl-D-aspartate receptor antago- nists on opiate tolerance and physical dependence. Neuropsychopharmacology 13:301-307, 1995 301. Trujillo KA, Akil H: Excitatory amino acids and drugs of abuse: a role for N- methyl-D-aspartate receptors in drug tolerance, sensitization and physical de- pendence. Drug Alcohol Depend 38:139-154, 1995 302. Trujillo KA, Akil H: Inhibition of morphine tolerance and dependence by the NMDA receptor antagonist MK-801. Science 251:85-87, 1991 303. Trujillo KA, Akil H: The NMDA receptor antagonist MK-801 increases mor- phine catalepsy and lethality. Pharmacol Biochem Behav 38:673-675, 1991 304. U'Prichard DC, Greenberg DA, Snyder SH: Binding characteristics of a radio- labeled agonist and antagonist at central nervous system alpha noradrenergic re- ceptors. Mol Pharmacol 13:454-473, 1977 305. Uchino S, Watanabe W, Nakamura T, et al: Establishment of CHO cell lines expressing four N-methyl-D-aspartate receptor subtypes and characterization of a novel antagonist PPDC. FEBS Lett 506:117-122, 2001 306. Ukai M, Maeda H, Nanya Y, et al: Beneficial effects of acute and repeated administrations of sigma receptor agonists on behavioral despair in mice ex- posed to tail suspension. Pharmacol Biochem Behav 61:247-252, 1998

74 307. Ulas J, Brunner LC, Geddes JW, et al: N-methyl-D-aspartate receptor complex in the hippocampus of elderly, normal individuals and those with Alzheimer's disease. Neuroscience 49:45-61, 1992 308. Ulas J, Cotman CW: Decreased expression of N-methyl-D-aspartate receptor 1 messenger RNA in select regions of Alzheimer brain. Neuroscience 79:973- 982, 1997 309. Urani A, Roman FJ, Phan VL, et al: The antidepressant-like effect induced by sigma(1)-receptor agonists and neuroactive steroids in mice submitted to the forced swimming test. J Pharmacol Exp Ther 298:1269-1279, 2001 310. Uzunova V, Sheline Y, Davis JM, et al: Increase in the cerebrospinal fluid content of neurosteroids in patients with unipolar major depression who are re- ceiving fluoxetine or fluvoxamine. Proc Natl Acad Sci U S A 95:3239-3244, 1998 311. Vajda FJ: Neuroprotection and neurodegenerative disease. J Clin Neurosci 9:4- 8, 2002 312. Watanabe M, Inoue Y, Sakimura K, et al: Developmental changes in distribu- tion of NMDA receptor channel subunit mRNAs. Neuroreport 3:1138-1140, 1992 313. Waterhouse RN, Chang RC, Atuehene N, et al: In vitro and in vivo binding of neuroactive steroids to the sigma-1 receptor as measured with the positron emis- sion tomography radioligand [18F]FPS. Synapse 61:540-546, 2007 314. Weaver CE, Jr., Marek P, Park-Chung M, et al: Neuroprotective activity of a new class of steroidal inhibitors of the N-methyl-D-aspartate receptor. Proc Natl Acad Sci U S A 94:10450-10454, 1997 315. Weaver CE, Land MB, Purdy RH, et al: Geometry and charge determine phar- macological effects of steroids on N-methyl-D-aspartate receptor-induced Ca(2+) accumulation and cell death. J Pharmacol Exp Ther 293:747-754, 2000 316. Weiland GA, Minneman KP, Molinoff PB: Thermodynamics of agonist and antagonist interactions with mammalian beta-adrenergic receptors. Mol Phar- macol 18:341-347, 1980 317. Weill-Engerer S, David JP, Sazdovitch V, et al: Neurosteroid quantification in human brain regions: comparison between Alzheimer's and nondemented pa- tients. J Clin Endocrinol Metab 87:5138-5143, 2002 318. Velisek L, Dreier JP, Stanton PK, et al: Lowering of extracellular pH suppresses low-Mg(2+)-induces in combined entorhinal cortex-hippocampal slices. Exp Brain Res 101:44-52, 1994 319. Vetulani J: Drug addiction. Part III. Pharmacotherapy of addiction. Pol J Pharmacol 53:415-434, 2001 320. Wihlback AC, Nyberg S, Andersson A, et al: Allopregnanolone serum concen- trations and neurosteroid sensitivity during withdrawal from postmenopausal hormone therapy. Gynecol Endocrinol:1-7, 2007 321. Williams K: Ifenprodil discriminates subtypes of the N-methyl-D-aspartate receptor: selectivity and mechanisms at recombinant heteromeric receptors. Mol Pharmacol 44:851-859, 1993 322. Williams K: Ifenprodil, a novel NMDA receptor antagonist: site and mechanism of action. Curr Drug Targets 2:285-298, 2001 323. Williams K: Modulation and block of ion channels: a new biology of poly- amines. Cell Signal 9:1-13, 1997 324. Williams K: Pharmacological properties of recombinant N-methyl-D-aspartate (NMDA) receptors containing the epsilon 4 (NR2D) subunit. Neurosci Lett 184:181-184, 1995

75 325. Williams K, Dawson VL, Romano C, et al: Characterization of polyamines having agonist, antagonist, and inverse agonist effects at the polyamine recogni- tion site of the NMDA receptor. Neuron 5:199-208, 1990 326. Williams K, Romano C, Dichter MA, et al: Modulation of the NMDA receptor by polyamines. Life Sci 48:469-498, 1991 327. Williams K, Russell SL, Shen YM, et al: Developmental switch in the expres- sion of NMDA receptors occurs in vivo and in vitro. Neuron 10:267-278, 1993 328. Vilner BJ, Bowen WD: Modulation of cellular calcium by sigma-2 receptors: release from intracellular stores in human SK-N-SH neuroblastoma cells. J Pharmacol Exp Ther 292:900-911, 2000 329. Vilner BJ, Bowen WD: Sigma receptor-active neuroleptics are cytotoxic to C6 glioma cells in culture. Eur J Pharmacol 244:199-201, 1993 330. Vilner BJ, de Costa BR, Bowen WD: Cytotoxic effects of sigma ligands: sigma receptor-mediated alterations in cellular morphology and viability. J Neurosci 15:117-134, 1995 331. Vilner BJ, John CS, Bowen WD: Sigma-1 and sigma-2 receptors are expressed in a wide variety of human and rodent tumor cell lines. Cancer Res 55:408-413, 1995 332. Wojtal K, Trojnar MK, Czuczwar SJ: Endogenous neuroprotective factors: neurosteroids. Pharmacol Rep 58:335-340, 2006 333. Wu FS, Gibbs TT, Farb DH: Pregnenolone sulfate: a positive allosteric modula- tor at the N-methyl-D-aspartate receptor. Mol Pharmacol 40:333-336, 1991 334. Wu G, Fang Y, Lu ZH, et al: Induction of axon-like and dendrite-like processes in neuroblastoma cells. J Neurocytol 27:1-14, 1998 335. Yan C, Hou Y: [Determination of neurosteroids in rat brain by gas chromatog- raphy/mass spectrometry]. Se Pu 22:12-15, 2004 336. Yesalis CE, Barsukiewicz CK, Kopstein AN, et al: Trends in anabolic- androgenic steroid use among adolescents. Arch Pediatr Adolesc Med 151:1197-1206, 1997 337. Young AB, Greenamyre JT, Hollingsworth Z, et al: NMDA receptor losses in putamen from patients with Huntington's disease. Science 241:981-983, 1988 338. Zheng F, Erreger K, Low CM, et al: Allosteric interaction between the amino terminal domain and the ligand binding domain of NR2A. Nat Neurosci 4:894- 901, 2001

76

Acta Universitatis Upsaliensis Digital Comprehensive Summaries of Uppsala Dissertations from the Faculty of Pharmacy 69

Editor: The Dean of the Faculty of Pharmacy

A doctoral dissertation from the Faculty of Pharmacy, 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 summary alone is distributed internationally through the series Digital Comprehensive Summaries of Uppsala Dissertations from the Faculty of Pharmacy. (Prior to January, 2005, the series was published under the title “Comprehensive Summaries of Uppsala Dissertations from the Faculty of Pharmacy”.)

ACTA UNIVERSITATIS UPSALIENSIS Distribution: publications.uu.se UPPSALA urn:nbn:se:uu:diva-8503 2008