Petar - University of Toronto

152
Direct Effects of Glucocorticoids and Serotonin on Developing Hippocampal Cells Petar Erdeljan A thesis submitted with the requirernents for the degree of Master of Science Graduate Department of Physiology University of Toronto O Copyright by Petar Erdeljan 2000

Transcript of Petar - University of Toronto

Page 1: Petar - University of Toronto

Direct Effects of Glucocorticoids and Serotonin on Developing

Hippocampal Cells

Petar Erdeljan

A thesis submitted with the requirernents for the degree of

Master of Science

Graduate Department of Physiology

University of Toronto

O Copyright by Petar Erdeljan 2000

Page 2: Petar - University of Toronto

National Libmry l*l d Canada Bibliothèque nationale du Canada

Acquisitions and Acquisitions et Bibliographie Services services bibliographiques 395 Wellington Street 395, rue Wellington Ottawa ON K I A ON4 ûttawa ON K1A ON4 Canada Canada

The author has graoted a non- L'auteur a accordé une licence non exclusive licence allowing the exclusive permettant à la National Library of Canada to Bibliothèque nationale du Canada de reproduce, loan, distribute or sell reproduire, prêter, disûibuer ou copies of this thesis in microfom, vendre des copies de cette thèse sous paper or electronic formats. la forme de microfiche/fïim, de

reproduction sur papier ou sur format électronique.

The author retains ownership of the L'auteur conserve la propriété du copyright in this thesis. Neither the droit d'auteur qui protège cette thèse. thesis nor substantial extracts f?om it Ni la thèse ni des extraits substantiels rnay be printed or otherwise de celle-ci ne doivent être imprimés reproduced without the author's ou autrement reproduits sans son permission. autorisation.

Page 3: Petar - University of Toronto

Direct Effects of Glucocorticoids and Serotonin on Developing

Hippocam pal Cells

Petar Erdeljan

MSc., 2000

Department of Physiology, University of Toronto

We hypothesized that dexamethasone. corticosterone/cortisol and

serotonin exposure wouid modify GR and MR mRNA expression within fetal

rnouse and fetal guinea-pig hippocampal ce11 cultures. In mouse cultures. 4 day

exposure to dexamethasone or corticosterone (IOnM or 100nM) down-regulated

levels of GR mRNA within neurons and exposure to serotonin (100nM)

significantly up-regulated expression of GR rnRNA. MR mRNA levels were

unaffected by any of the treatments. Dexamethasone, corticosterone or serotonin

exposure did not alter expression of GR mRNA within glial cells. In guinea pig

cultures, 4 day exposure to dexamethasone or cortisol (1 00nM) down-regulated

levels of GR mRNA within neurons and exposure to serotonin (100nM)

significantly up-regulated expression of GR mRNA. Finally, multidrug resistance

(MDRla) gene expression excludes dexamethasone from the brain. We found

that the MDRla gene is expressed in the hippocampus. dentate gynis. specific

thalamic nuclei and cortex of the fetal brain. Patterns of MDRla changed dunng

developrnent.

Page 4: Petar - University of Toronto

ACKNOWLEDGEMENTS

I whish to thank the following individuals:

Dr. Stephen G. Matthews: For being my first mentor in science. Through Our

friendship I became aware of new dimensions in thinking and approach to

science. For his supervision. guidance, support dumg my studies. i whish to

thank him for his patience and for allowing me to express my sometimes "radicaln

creativity .

Dr. John F. MacDonald: For allowing me to work in his laboratory and for his

expert scientific input into rny project.

Dr. Denise Belsham: For being a member of my supetvisory committee and for

her suggestions and advice.

Elrzbieta Czewinska: For her

this study.

would have

Li Liu and

Without her expert

been impossible.

help in developing the culture system utilized in

advice, suggestions and patience this project

Lucy McCabe, who ensured cuntinuous support of experimental

animais used in this study.

Rania Lingas and Joanne Kotsopoulos who provided friendly and very

enjoyable atmosphere in the lab.

My parents for their endless support and encouragement.

Page 5: Petar - University of Toronto
Page 6: Petar - University of Toronto

CHAPTER 3

Regulation of GR and MR mRNA during Development: Mouse

Studies. ............................................................................................. 34

3.1 Introduction .................................................................................... 34

..................................................................................... 3.2 Objectives 35

3.3 Hypothesis ..................................................................................... 36

3.4 Methods ....................................................................................... -36

3.4.1 Mouse Hippocampal Culture ............................................................ 36

3.4.2 In situ Hybridization ....................................................................... 38

3.4.3 Oligonucleotide Probes ................................................................... 38

3.4.4 Probe Labeling ............................................................................. 38

.............................................................................. 3.4.5 Hybridization -39

3.4.6 Washing ...................................................................................... 40

3.4.7 Emulsions ................................................................................... 40

3.4.8 Developing and Staining Emulsions .................................................. 41 . *

3.4.9 Cell Viability ................................................................................. 41

3.5 Data analysis .................................................................................. 42

.......................................................................................... 3.6 Results 43

..................................................................................... 3.7 Discussion 45

CHAPTER 4

Reguiation of GR and MR mRNA During Development: Guinea Pig Studies.

Gestational Age 40 Days ............................................~....................... ..61

Introduction .................................................................................... 61

Objectives .................................................................................... -63 Hypothesis .................................................................................... -63

Methods ....................................................................................... -64

Development of Fetal Guinea-pig Culture ........................................... 64

Mouse Cerebellar Culture ............................................................... 65

Guinea-pig Hippocampal Culture, Gestational Day 40 .......................... 65

Page 7: Petar - University of Toronto

4.4.4 In situ Hybridization ....................................................................... 68

.................................................................. 4.4.5 Oligonucleotide Probes 68

............................................................................... 4.4.6 Hybridization -68

...................... 4.4.7 Washing, Emulsions. Developing and Staining Emulsions 68

............................................................................... 4.5 Data Analysis 69

......................................................................................... 4.6 ResuIts -70

.................................................................................... 4.7 Discussion -71

CHAPTER 5

Regulation of GR and MR mRNA During Development: Guinea Pig Studies.

Gestational Age 50 Dayç: .........................~......~.......~...m...~..~.........~.... **a3

.................................................................................... Introduction 83

.................................................................................... Objectives -83

.................................................................................... Hypothesis -84

........................................................................................ Methods 85

Development of Fetal Guinea-pig Culture system ................................. 85

Mouse cerebellar culture on inserts ....................... ,... ................... 85

Guinea-pig hippocampal culture. gestational day 50 ......................... ... . 86 ...................................................................... In situ hybridization -86

.................................................................................. 5.5 Data analysis 86

.......................................................................................... 5.6 Results 87

..................................................................................... 5.7 Discussion 88

CHAPTER 6: Multi drug resistance studies ...................................m.......m 95

.................................................................................... 6.1 Introduction 95

.................................................................................... 6.2 Objectives -96

..................................................................................... 6.3 Hypothesis 96

....................................................................................... 6.4 Methods -97

............................................ ......... ............... 6.4.1 Animal Work ,. ,,. 97

6.4.2 Tissue Preparation and Sectioning ................... .... .......................... 97

6.4.3 ln situ Hybridizaüon ...................................................................... 98

Page 8: Petar - University of Toronto

........................................................... 6.5 Autoradiography and Analysis -98

.......................................................................................... 6.6 Results 99

................................................................................... 6.7 Discussion 701

CHAPTER 7: Summaw and Conclusions ........m..............m.m.....m......~...... 114

APPENDlX 1: Culture solutions ..............m........m.........................m........ 118

APPENDIX 2: Derivation of treatments for mouse hippocampal cultures.120

.......................................... APPENDIX 3: Molecular biology solutions A22

REFERENCESmemm*eeem..... ............................................................. ..l28

Page 9: Petar - University of Toronto

LIST OF FIGURES

Figure 3.la: Mouse hippocampal neurons in vitro and . . ................................................... autoradiographic images.. .51

Figure 3.1 b: Silver emulsion grains over mouse hippocampal neurons.. ......... 53

Figure 3.2a: Analysis of GR mRNA in the mouse neurons following

................................................. dexamethasone treatment.. .55

Figure 3.2b: Analysis of MR mRNA in the mouse neurons following

dexamethasone treatment.. ................................................. .55 Figure 3.3a: Analysis of GR mRNA in the mouse neurons following

corticosterone treatment, .................................................... .57

Figure 3.3b: Analysis of MR mRNA in the mouse neurons following

corticosterone treatment. ..................................................... .57

Figure 3.4a: Analysis of GR mRNA in the mouse neurons following

serotonin treatrnent,. ......................................................... .59 Figure 3.4b: Analysis of MR mRNA in the mouse neurons following

.......................................................... Serotonin treatment.. .59

Figure 4.la: Analysis of GR mRNA in the guinea-pig neurons following

corticosterone treatment, .................................................... .80

Figure 4.1 b: Analysis of MR mRNA in the guinea-pig neurons following

corticosterone treatment.. ................................................... .80

Figure 4.2: Analysis of GR mRNA in the guinea-pig neurons following

dexamethasone treatmenf.. ............................,.................... .82

Figure 4.3: Analysis of GR mRNA in the guinea pig neurons following

serotonin treatment.. .......................................................... .82

Figure Sala: Analysis of GR mRNA in the guinea-pig neurons (gd50) following

dexamethasone exposure.. ................................................. -92 Figure 5.l b: Analysis of MR mRNA in the guinea-pig neurons (gd50) following

dexamethasone exposure.. ................................................ -92 Figure 5.2a: Analysis of GR mRNA in the guinea-pig neurons (gd50) foliowing

serotonin exposure.. .......................... .. ......................... ..94

Page 10: Petar - University of Toronto

Figure 5m2b: Analysis of MR mRNA in the guinea-pig neurons (gd50) following

Figure 6.1:

Figure 6.2:

Figure 6.3:

Figure 6.4:

Figure 6.5:

Figure 6.6:

Figure 6.7:

Figure 6.8:

Figure 6.9:

Table 1 :

serotonin exposure.. ........................................................... 94

Color-enhanced image of MDRI a in coronal section of a fetal

............................................................ guinea-pig brain.. -1 05

Images of coronal sections that underwent silver emulsion

............................................................. autoradiography. .IO5

MDRla mRNA levels within the CAIICA.2 region of the

................................................................ hippocampus.. -1 07

MDRla mRNA levels within the CA3 region of the

Hippocampus.. ............................................................... -1 07

MDRl a mRNA levels within the CA4 region of the

................................................................ hippocampus.. .l 09

MDRl a mRNA levels within the dentate gyms.. ....................... 109

MDRl a mRNA levels within the cortex.. ............................... .l Il M DR? a mRNA levels within the thalamic nuclei ...................... . I l 1

MDRla mRNA levels in the limbic regions of fetal guinea-pig

following dexamethasone exposure.. .................................. .Il 3

Effects of dexarnethasone, corticosterone and serotonin on GR

mRNA expression in non-neuronal cells.. .............................. .60

Page 11: Petar - University of Toronto

CHAPTER 1 : Introduction

1 .el Overview

Fetal glucocorticoid exposure or prenatal stress can permanently modify

the activity of the h ypothaiamic-pituitary-adrenai (HPA) axis. This modification or

"programmingn is at least in part mediated by an alteration in expression of

corticosteroid receptor genes in the brain reg ions involved in g l ucocorticoid

feedback (Matthews, 2000). Evidence from animal models indicated that the

hippocampus, a key locus for glucocorticoid feedback control, is central to the

process of programming (Jacobson and Sapolsky, 1991).

Permanent modification of the HPA axis is associated with alterations in

basal and stress-induced glucocorticoid secretion. Studies in rats demonstrated

that elevation of circulating glucocorücoid is linked to a decrease in hippocarnpal

glucocorticoid receptor levels, following prenatal dexamethasone (a synthetic

glucocorticoid) administration (Levitt et al., 1996). lncreases in the basal and

stress-induced levels of g lucocorticoid were also reported in the rhesus mon key

following prenatal synthetic glucocorticoid administration (Uno et al. 1994). Such

treatment is known to result in degeneration and depletion of pyramidal neuron

cells and granular neurons in the hippocampus, as well as in an overall reduction

in the hippocampal size and segmental volume (Uno et al. 1990). Unfortunately,

studies done in rhesus monkeys did not address potential changes in

corticosteroid receptor levels within the hippocarnpus.

Page 12: Petar - University of Toronto

Chronic increases in circulating glucocorticoid have been associated with

increased susceptibility to pathologies such as depression, hypertension,

immunosupression and diabetes (Baxter et al., 1987; Munck et al., 1984;

Sapolsky, 1994). Development of hypertension is strongly associated with

prenatal alteration of HPA function, at least in the rat model. In the rat, prenatal

dexamethasone treatment results in the adult offspring having elevated blood

pressure (BenediMsson et al., 1993). At present, the precise mechanisms

underlying the development of hypertension are unknown, as is the link between

hypertension, elevated glucocorticoids and reduction in the hippocampal

glucocorticoid receptor levels.

At present, 10% of al1 women, who are at nsk of pre-terni delivery are

treated with synthetic glucocorticoids. Synthetic glucocorticoid therapy is

administered in order to promote organ maturation and thus avoid complications

associated wit h pre-term delivery, such as respiratory distress syndrome,

intraventrîcular hemorrhage and necrotizing colitis (NIH consensus development

conference, 1995). Synthetic glucocorticoid therapy is used extensively and

often in a repeated fashion (French, et al. 1997). However, long-terrn effects of

synthetic glucocorticoid therapy on brain development (in the human) are not

known. To date, there is little information on the possible direct effects of

glucocorticoid exposure on expression of corticosteroid receptors in fetal

hippocampal neurons. The primary purpose of this study was to examine this

issue.

Page 13: Petar - University of Toronto

1.2 Stress and the Hypothalarno-Pituitary-Adrenal (HPA) axis

The brain can interpret a wide range of stress-related inputs into a general

class of neuroendocrine and autonomic responses designed to allow the

organisrn to cope with stress. The hypothalamic-pituitary-adrenal (HPA) systern

represents a principal component of the stress response. whose primary role is

to allow and to control secretion of glucocorticoids (GCs) by the adrenal cortex.

Glucocorticoid actions are diverse and wiil be considered in detail later.

The primary role of the stress response (particulariy glucocorticoids) is to

facilitate adaptation to stress and to restore homeostasis. Neurons in the

paraventrîcular nucleus (PVN) of the anterior hypothalamus are essential to the

stress response. Neurons within the PVN synthesize and secrete corticotrophin

release hormone (CRH), its CO-secretagogue vasopressin (VP) as well as other

neuropeptides that drive the activity of the sympatho-medullary systems. The

sympatho-medullary and HPA system act in a CO-ordinated fashion and exert

reciprocal control over the other activity (De Kloet, et al., 1998).

Upon stimulation from higher or lower brain centers, specialized

neurosecretory cells in the PVN release CRH, VP and oxytocin (Whitnall, 1993;

Plotsky, 1989). These hypophysiotrophic factors are released from nerve

teminals in the zona extema of the median eminence into the primary plexus of

the hypophyseal portal system (Antoni, 1986; Plotsky, 1987). Primary plexus

capillaries merge into the long portal vessels that lead to the infundibular stalk of

the pituitary gland (Ambrach et al., 1976). The released hypophysiotrophic

factors stimulate the synthesis and release of adrenocorticotrophin releasing

Page 14: Petar - University of Toronto

hormone (ACTH) from corücotroph cells in the anterior pituitary (Plotsky, 1987).

ACTH is foned from proopiomelanocortin (POMC) which is processed into

ACTHi-39, pendorphin, p-lipotrophin within corticotroph cells. In intermediate

pituitary melanotroph cells, a-MSH, CLIP and acetylated P-endorphin are primary

products of POMC processing (Eipper and Mains, 1980). ACTH, once released,

enters the systemic circulation and acts on adrenocortical cells to initiate

synthesis and secretion of glucocorticoids (Plotsky, 1989).

Release of glucocorticoids allows the organism to cope with, adapt to, and

recover from stress. Traditionally, glucocorticoids have been thought to act as a

prirnary defensive mechanism, responsible for initiation of physiological actions

needed for immediate survival. In addition, glucocorticoids also have the potential

to initiate a number of adaptive responses requiring genomic activation. lnside

the cell, glucocorticoids bind to high affinity cytosolic receptor and act via

genomic mechanisms to modiFy function (Munck, et al. 1984; Bamberger, et al.

1996). However, it has also been suggested that glucocorticoids may also act on

a membrane bound receptor and in this way rapidly alter cellular events (Brann,

et al. 1995; Bamberger, et al. 1996).

The actions of glucocorticoids display two modes. In the "proactive" mode

glucocorticoids maintain basal activity of the H PA system and control the

sensitivity of the systems to stress. Glucocorticoids promote CO-ordination of

circadian events, such as food intake and sleepiwake cycle, and are involved in

processes underlying selective attention, integration of sensory information and

response selection. In "reactiven mode glucocorticoid feedback teminates stress-

Page 15: Petar - University of Toronto

induced HPA activation (De Kloet. et al. 1998), preventing excessive

glucocorticoid exposure which is detrimental to health (Munck, et al. 1 984;

Munck, et al. 1992; Tausk, 1951). It has been known for several decades that

lack of wntrol over the stress response can increase vulnerability to disease

(Seyle, 1952).

1.3 Corticosteroid Receptors and the Hippocampus

Glucocorticoids act on the brain through glucocorticoid receptors (GRs)

and mineralocorticoid receptors (MRs) (McEwen et al, 1986; De Kloet, 1991).

GRs are present throughout the brain but are rnost abundant in hippocampus,

hypothalamic CRH neurons and pituitary corticotrophes. MRs are present at

hypothalamic sites involved in regulation of salt appetite and autonornic

regulation (Broody, 1980; McEwen, 1986; Gomez-Sanchez, 1997). However, the

highest concentration of MRs is detected within the hippocampus, a brain

structure involved in leaming and memory processes. In the hippocampus

corticosterone binds both MR and GR. This is in contrast to the situation in the

perip heral organs. In the kidney, M Rs selectively bind the mineralocorticoid

aldosterone despite the fact corticosterone can bind MRs with equal or greater

affmity than aldosterone and that its circulating concentration is up to 1000-times

greater than that of aldosterone (Seckl. 1997). In the hippocampus the

mineralocorticoid (ie aldosterone) selectivity for MR is lost (Korozowski, 1983;

Reul, 1985; McEwen, 1968; De Kloet, 1975). The selectivity in the kidney is

controlled by the enzyme 1 1 P-hydroxysteroid dehydrogenase, type2 (1 1 p-HSD2).

Page 16: Petar - University of Toronto

This enzyme, which is present primarily in the placenta and kidney, deactivates

glucocorticoids by conversion to their inactive metabolites (Ajilore and Sapolsky,

1998). Steroid regulation in the brain is thought to involve 11P-HSDI. This more

ubiquitous isoform. is present in hypothalamus, hippocampus, neocortex and

subthalamus (Moisan, et al. 1992, Lakshmi, et al. 1991 ; Seckl, et al. 1991). 1 1 P-

HSD1 is bidirectional, and thus. it could either act as a dehydrogenase (to

deactivate glucocorticoids) or as a reductase (to regenerate glucocorticoids). At

present, it is believed that 11P-HSD1 acts primarily as a reductase (Ajilore and

Seckl, 1998), thus converting inactive metabolites to active corticosterone. This

speculation rernains somewhat controversial. In the hippocampus, MRs

(Kd-O.5nM) bind glucocorticoids with approximately 10-fold higher affinity than

GRs (Kd-5nM) (Korozowski, 1983; Reul, 1985; McEwen, 1968; De Kloet, 1975).

Under conditions of basal corticosterone plasma levels, MRs will be primarily

occupied, while GRs becorne occupied with increasing concentrations of

corticosterone in the plasma (De Kloet, et al. 1998). Hence, in the hippocampus,

glucocorticoids activate two signaling pathways via MRs and GRs (Reul, 1985).

Occupation of GRs and MRs will have profound effects on neuron activity, some

of which are described below.

Activation of MRs and GRs is associated with modification of neural firing.

Activation of MRs in CA1 hippocampal neurons is associated with small, voltage-

gated calcium currents. Activation of GRs is associated with a n'se in calcium

current amplitude (Karst et al., 1994). GR mediated effects were found to be

dependent on de novo protein synthesis (Kerr et al., 1992) and they affected both

Page 17: Petar - University of Toronto

basal and stimulus-induced intracellular calcium levels (Elliot and Sapolsky,

1 992; Elliot and Sapolsky. 1 993). Such alterations in intracellular calcium

homeostasis affected the activation of a slow calcium-dependent potassium-

channels (Storm. 1990). This hyperpolarizes neurons after prolonged firinig.

Hyperpolarization of neurons results in reduction of firing (accommodation) (De

Kloet, et al. 1998). Neural firing is also suppressed by afterhyperpolarization (De

Kloet, et al. 1998). Predominant MR activation was found to result in small

accommodation and after-hyperpolarization amplitude. Additional occupation of

GRs was found to increase accommodation and afterhyperpolarization amplitude

(Joels and De Kloet, 1990). In summary, CA1 hippocampal output to the PVN

neurons is effectively increased with MR activation (at basal corticosterone

levels) and it is reduced when both GR and MR are activated (at stress

corticosterone levels).

Glutamate-mediated responses are also affected by MR and GR

activation. Glutamate is the predominant transmitter molecule in the CA1 area of

the hippocampus. Glucowrticoid activation of MRs is associated with no change

in excitatory transmission, w hile glucocorticoid activation of G Rs is associated

with the depression of glutarnatergic transmission (Joels and Fernhout, 1993).

Additionally, slow inhibitory postsynaptic potentials. mediated by GABAb

receptors, remain unchanged when MRs are activated. GR activation is found to

suppiess these slow inhibitory potentials (De Kloet et al., 1998). Fast inhibitory

postsynaptic potentials are not greatly affected by changes in MWGR activation

(De Kloet et al., 1998). In summary, both excitatory and inhibitory information

Page 18: Petar - University of Toronto

rernains unchanged under MR activation. Additional GR activation under

elevated glucocorticoid levels reduces excitatory transmission and hence

negative CA1 hippocampal output to the PVN (De Kloet et al., 1998).

Activation of GRs and MRs also modifies actions of biogenic amines,

noradrenaline and serotonin, in the hippocarnpus. Slow calcium-dependent

potassium conductance is blocked by no rad renaline, via P-ad renergic receptor.

Inhibition of this curent results in increased excitability of the CA1 hippocampal

area (Madison and Nicoll, 1986). Activation of GRs is associated with a reduction

of noradrenaline effect, and hence a decrease in CA1 excitability (De Kloet et al.,

1998). Effects of MR activation are still unknown.

Serotonin has many different effects in the CA1 hippocampal area. One of

the most prominent effects of serotonin is hyperpolarization of the CA1 neurons.

This hyperpolarkation of the membrane is achieved through 5HTla receptors

(Andrade and Nicoll, 1997). Activation of MRs is associated with srnall

afterhyperpolan'zation, whereas additional GR activation increases serotonin-

induced hyperpolarization (Joels and De Kloet, 1 992). The inhibitory effect of

serotonin under conditions of GR activation results in a marked suppression of

excitatory transmission in the CA1 area (Hesen et al., 1998).

1.4 GR and MR Transactivation

lntracellular GRs and MRs are part of a cytoplasmic multiprotein complex.

This complex consists of one receptor molecule. hnro molecules of heat shock

protein (hsp) 90, one hsp70, one hsp56 and one immunophilin molecule (Smith

Page 19: Petar - University of Toronto

and Toft, 1993). Activation of the corticosteroid receptors results in rapid

dissociation of this molecular complex, phosphorylation of the receptor at multiple

sites and increased affinity of the ligand-activated receptor for nuclear domains

(Brink, et al. 1992). Activated corticosteroid receptor homodimerizes and

translocates to the nucleus where it interacts with specific DNA sequences or

other transcription factors (Barnberger. et al. 1996). Binding of GRs to a specific

DNA sequence represents the classic model of corticosteroid action. The

receptor homodimer binds to a glucocorticoid response element (GRE), which is

a short palindromic DNA sequence in the promoter region of the glucocorticoid

responsive genes. Once bound to the GRE. the GR homodimer interacts directly

with members of the basic transcription machinery, such as TFIIP. Indirect

interaction via bridging factors, such as steroid receptor coactivator 1 (SRCI), is

also possible. Recent evidence indicates that GRs can act by recruiting multiple

wregulator proteins that may have specific functions during transcriptional

initiation. Activated receptors mobilize members of the SRC family, which is a

group of stnicturally and functionally related transcriptional coactivators.

Interaction with transcriptional cointegrators p300 and CPB is also implicated in

the GR activation. P300 and CPB are proposed to integrate diverse afferent

signals at hormone-regulated promotors (McKenna et al. 1999). Other nuclear

receptor coactivators include BRGI (it couples ATP-hydrolysis to chromatin

rernodeling) and the E3 ubiquitin-protein ligases E6AP and RPFI. Any of the

mentioned interactions (direct or indirect) are sufficient to stabilize the pre-

initiation complex on the promoter and, therefore, enhance transcription by RNA

Page 20: Petar - University of Toronto

polymerase II (Barnberger, et al. 1996). In addition, the activated GR can also

bind to negative glucocorticoid response elements. Genes that do not contain

GREs can also be affected by GR activation. One way in which this can be

accomplished by the GR-mediated regulation of transcription factors. These two

mechanisms will be considered in detail. later.

Rapid, short duration effects of GCs are thought to be mediated by

signaling via a membrane bound glucocorticoid receptor (Hua and Chen, 1989).

At present, such a receptor has not been isolated or characterized. though its

existence has been demonstrated indirectly (Hua and Chen, 1989). Within 2

minutes of their application. glucocorticoids can h yperpolarize the membrane

potential of hippocampal neurons in brain slices, and coeliac ganglion neurons.

Bovine aibumin-glucocorticoid conjugates, which cannot enter the cell, produce

the same effect (Hua and Chen, 1989). Considering this finding and the rapidity

of effect, existence of membrane bound receptor is likely.

1.5 The Hippocampus and Regulation of HPA Function

Considerable evidence indicates that the hippocampus inhibits HPA

activity. lnhibitory hippocampal GABA-ergic projections to the hypothalamus

were identified and characterized (Nauta. 1956; Mason. 1958; Herman, 1996).

Electrical stimulation of hippocampal sub-fields CA1 (unilateral) and subiculum in

humans resulted in reduction of 47-hydroxy corticoids (cortisol metabolites) in the

plasma (Rubin et al, 1966). Electrical stimulation of hippocampal sub-fields CA3,

CA4, the dentate gyrus and subiculum significantly reduced stress-stimulated

Page 21: Petar - University of Toronto

plasma corticosterone levels (Dunn and Or , 1984). Taken together these studies

suggest that the hippocampus is involved in tonic inhibition of HPA function.

Experiments utilizing hippocampectomy or hippocampal neuron

destruction also support an inhibitory role of the hippocampus. Destruction of

hippocamapal neurons (>50%) by sterotaxic kainate infusion increased plasma

corticosterone levels during restraint stress when compared to control. in rats

(Sapolsky et al. 1984). Total hippocampectomy of the dorsal hippocampus in the

rat resulted in a 4-fold increase in CRH mRNA expression in the PVN relative to

sham-operated animais (Herman et al, 1989). This study confined that the

inhibitory effects of the hippocampus on pituitary adrenocortical function are

mediated via the hypothalamus. Furthemore, hippocampectomy in the rat leads

to an increase in basal and stress-induced levels of ACTH, when wmpared to

sham-controls (Wilson et al, 1980). This finding also confimis that the

hippocampus in hi bits ACTH release. Finally, partial hippocampectomy (only

dorsal but not ventral), in the rat reduced the inhibitory effect of dexamethasone

on basal and stress-induced adrenocortical response (Feldman et al, 1 980). This

study suggests that dorsal hippocampal formation participates in feedback

regulation of the HPA function.

The hippocampus influences the release of adrenaline from brainstem

catecholaminergic nuclei, which innervate the parvocellular field of the PVN

(Meibach, 1977; Swanson, 1977, 1981). This finding suggests that the

hippocampus indirectly infiuences the neurosecretory activity of parvocellular

neurons which directly control ACTH release (Swanson, 1977, 1981 ).

Page 22: Petar - University of Toronto

The role of hippocarnpal MRs and GRs in HPA regulation was elegantly

demonstrated by a series of experiments utilizing selective receptor antagonists

in adrenally intact rats. lntraventricular administration of MR antagonist was

demonstrated to increase basal levels of plasma corticosterone as well as stress-

induced levels of corticosterone (Ratka et al, 1989). This effect was observed

both in the moming and the aftemoon phase (in rats, highest levels of

corticosterone are observed in the afternoon phase and lowest corticosterone

levels are observed in the moming phase). Similar observations were made in

the human. Systemic administration of spironolactone (MR antagonist) was found

to increase basal HPA activity (Dodt et al, 1993), though this response was not

noted in al1 studies (Michelson et al.. 1994). Together the data would suggest

that hippocampal MRs mediate the effect of corticosterone in maintaining the

tone of basal HPA activity.

lntraventricular administration of RU38486 to rats (GR antagonist) had no

effect on basal levels of corticosterone in the rnoming phase (Ratka et al, 1989).

lntraventncular RU38486 administration in the aftemoon phase increased HPA

activity. In contrast, administration of the same antagonist directly into the

hippocampus decreased ACTH levels in the aftemoon phase (Van Haarst et al.,

1997). These contradictory findings suggest that hippocampal GRs are prïmarily

involved in decreasing the inhibitory hippocampal outfiow to the PVN (De Kloet et

al, 1998). These findings also indicate that extra-hippocampal GRs, expressed in

PVN neurons, suppress the HPA activity.

Page 23: Petar - University of Toronto

In sumrnary, emerging evidence suggests that GR and MR have opposite

roles, with respect to HPA control, at the level of the hippocampus. This

correlates well with the data underiying MR and GR activation, which was

introduced previously. In this scenario. predominant MR activation maintains

hippocampal excitability, and inhibitory polysynaptic projections to the PVN, in

tum, suppress basal HPA activity. However activation of GRs suppresses the

hippocampal output and results in disinhibition of PVN neurons and in increase of

the HPA activity (De Kloet. et al. 1998). The hippocampal input seems to be

overridden at the level of PVN (at elevated corticosterone levels). This is a

relatively new hypothesis and it requires further testing.

1.6 Programming of the HPA Axis by Excess Glucocorticoids

Glucocorticoids are critical for normal brain developrnent, as they exert a

wide range of effects in most regions of the growing brain. Their presence is

necessary for many processes ranging from subcellular reorganization to neuron-

neuron and neuronglia interaction (Gould. 1994; Gould et al, 1991). Predictably,

their absence or prolonged elevation in the fetal brain is detrimental to normal

development and can permanently modty the structure and function of the brain

(Oppenheimer et al. 1997; Bohn. 1984).

It is well established that HPA function can be permanently programmed

during development (Levine, 1957). Available evidence in animal models

indicates that prenatal exposure to glucocorticoids can lead to offspring with

altered HPA activity. In the rat, fetal exposure to synthetic glucocorticoid,

Page 24: Petar - University of Toronto

dexamethasone (O.lrnglkg), during the last week of gestation resulted in adult

offspring with elevated basal plasma corticosterone levels (Levitt et al, 1997).

Dexamethasone exposed offspring were also found to have increased blood

pressure. Similarty, dexamethasone treatment (0.05rngIkg) on days I ï , l 8 and 19

of pregnancy resulted in adult offspring which had increased stress-induced

corticosterone levels (Muneoka et al, 1997). Treatment (0.4mglkg) on days 17

and 19 alone was not sufficient to alter HPA function in offspring. However, the

same animals were found to have a difference in the ratio of AVP and CRH in the

extemal zone of the median eminence, suggesting subtle long-term effects on

HPA control (Bakker et al, 1995). These studies demonstrate that glucocorticoids

can program the HPA axis, and that the nature of such programming is

dependent on dose and timing of exposure.

In the guinea-pig, fetal dexamethasone exposure (1 mglkg) on days 50

and 51 of gestation (75% of gestation) resulted in male offspnng (neonate) with

elevated (2-fold) basal plasma cortisol concentrations (Dean et al, 1999).

Similarly, fetal dexamethasone exposure (1 mglkg) on days 40 and 41, 50 and

51, 60 and 61, resulted in fernale offspring (adult) with elevated basal plasma

cortisol concentrations. However, the sarne treatment produced male offspring

with reduced basal plasma cortisol concentrations indicating a sex-specific

influence of exogenous glucocorticoids (Matthews, et al. 2000). In rhesus

monkeys, daily administration of dexamethasone (4 x 1.25mglkg) starting on

132d of gestation resulted in the young offspring with significantly elevated basal

and stress-induced levels of cortisol (Uno et al, 1994). These three studies

Page 25: Petar - University of Toronto

indicate that HPA function can be permanently altered in species that give birth

to neuroanatomically mature young (guinea-pigs and rhesus monkeys), a

situation similar to the human. To date, no studies have examined HPA function

in humans that were exposed to antenatal glucocorticoids during fetal life.

lncreased HPA activity throughout life will elevate tissue exposure to

g lucocorticoids. In humans, elevated tissue exposure to cortisol has been

associated with increased arteriosclerosis, cholesterol concentrations, incidence

of diabetes, immunosupression and cognitive impairment (Lupien et al, 1997;

Brindley et al, 1989; Munck et al. 1994). Similar effects of permanently altered

HPA function have been described in animal studies (Ader et al, 1973). In

contrast, long-ten reduction in HPA activity could have a protective effect

against cortisol tissue over-exposure effects.

HPA programming occurs, at least partially, at the level of the

hippocampus. The hippocampus is very sensitive to endogenous and exogenous

gluwcoricoids during development (Takahashi, 1998; Weinstock, 1997; De Kloet

et al, 1998; Matthews. 2000).

1.6.1 Programming of Hippocampal GRs and MRs by Excess

Glucocorticoid

The hippocampus wntains the highest levels of corticosteroid receptor

observed in any brain region (McEwen et al, 1968; Grlach et al, 1976; Reul et al,

1985; Sutanto and De Kloet. 1987; Matthews, 1998). It has been postulated that

the hippocampus exerts its input to the HPA axis primarily by changing the

Page 26: Petar - University of Toronto

sensitivity of glucocorticoid negative feedback (Motta et al, 1970; Jacobson et al,

1991). In support of this staternent, it has been recorded that the loss of

hippocampal corticosteroid receptors disrupts the sensitivity of the HPA axis to

glucocorticoid feed back in hi bition (Sapolsky et al, 1986; Levitt, et al. 1996).

At basal plasma glucocorticoid concentration hippocarnpal MR is occupied

(at approximately 80%) while the GR remains largely unoccupied. This is due to

the higher affinity of the MR for glucocorticoid (De Kloet et al, 1998). However.

during stress-induced elevation of glucocorticoid the GRs become increasingly

occupied (Dallman et al, 1994). Based on these findings it has been postulated

that the MR is primarily involved in feedback regulation during basal secretion of

stress hormone, while the GR is involved in regulation durhg increased

glucocorticoid secretion (Dallman et al, 1994). According to such a postulate, GR

activation is necessary for an increase in inhibitory hippocampal output to the

PVN, which is required to restore the HPA axis to basal state. However, as

described eariier, this view has been recently challenged. According, to De Kloet

and colleagues, GR activation is linked with an increase in HPA activity by

removal of inhibitory output from the hippocampus to the PVN. Under conditions

of GR activation hippocampal neurons were found to be less excitable, their

afterhyperpolarization state longer and their firing frequency lower (De Kloet et al,

1998), al1 of which promote a decrease in hippocampal output. However, this

remains to be tested further. Based on either theory, one can predict that

permanent change in the hippocampal corticosteroid receptor complement wiil

profoundly affect hippocampal inhibitory output to the PVN.

Page 27: Petar - University of Toronto

Evidence for a permanent change in corticosteroid receptor complement,

within the hippocarnpus, is provided by animal studies of fetal dexamethasone

exposure. In the rat, daily dexamethasone administration during the last week of

pregnancy reduced concentrations of both GR and MR mRNA in the

hippocampus (Levitt et al, 1996). There was no difference in GR mRNA or MR

mRNA expression in the hypothalamus or in brainstem structures. This reduction

in corticosteroid receptor mRNA was associated with an elevation of basal HPA

act ivity.

Studies undertaken in the guinea pig revealed that prenatal

dexamethasone exposure, during the phase of rapid brain growth (gestational

days 50 and 51 ), results in offspring with an altered hippocarnpal corticoseroid

receptor complement (Dean et al, 2000). Male offspring neonates were found to

have increased GR mRNA levels in the cingulate cortex and hippocampal sub-

field CA3. In wntrast, fernale neonates were found to have decreased GR mRNA

complement in the hippocarnpal sub-fields CA3 and CA4, dentate gyrus and

cingulate cortex (Dean et al, 2000). There was no effect of prenatal

dexamethasone exposure on GR mRNA concentrations in the hypothalamus and

anterior pituitary in either sex. Elevation of GR mRNA levels within the

hippocampus, in the male neonates, suggest an increase in inhibition of

hippocarnpal neurons, and thus, a decrease in the inhibitory tone to the PVN (De

Kloet et al, 1998). This correlates well with the increase in male basal plasma

cortisol concentrations. In the females. the situation is not as clear, as decreases

in the hippocampal GR mRNA did not result in a decrease in basal plasma

Page 28: Petar - University of Toronto

cortisol concentrations. Fetal guinea-pig glucocorticoid exposure on gestational

days 40 and 41, 50 and 51, 60 and 61, also results in alteration of hippocarnpal

corticosteroid receptor complement, in adult offspring (Matthews, et al. 2000).

Female adult animals (80 days) were found to have decreased MR mRNA levels

throughout the hippocampus. There were no differences in GR mRNA levels in

the hippocampus, but GR mRNA levels were decreased in the PVN and pituitary.

Male adult animals showed no difference in either GR mRNA or MR mRNA, at

the level of the hippocampus, PVN or pituitary (Matthews, et al. 2000). In adult

females, the decrease in MR mRNA suggests a decrease in inhibitory tone from

the hippocampus. Decreases in GR mRNA at the level of the PVN and the

pituitary suggest a decrease in negative feedback at these sites. 60th of these

speculations correlate well with the increase in the HPA function reported in

female adult animals (Matthews, et al. 2000). The situation in the adult males,

which were reported to have decreased HPA activity. is more difficult to

speculate upon. It is possible that GC exposure in utero has an impact on some

other brain site involved in the HPA regulation or affects other parameters rather

than the GRfMR mRNA expression.

Very little is known about the mechanisms that underlie long-terni

alteration of corticosteroid receptor densities. It is possible that glucocorticoids

can influence the pattern of the GR and MR developrnent at the time of

exposure. In the rat, GR mRNA can be detected in the hippocampus by

gestational day 13 and MR mRNA by gestational day 16 (Cintra et al, 1993; Diaz

et al, 1998). In general, bath GR and MR levels in the rat brain are low

Page 29: Petar - University of Toronto

throughout gestation and then increase rapidly after birth (Bohn et al, 1994;

Rosenfeld et al, 1993; Meaney et al, 1985; van Eekelen et al, 1991). In the fetal

guinea-pig, both GR mRNA and MR mRNA can be detected in the hippocampus

by gestational day 40 (Matthews, 1998). Between gestational days 40 and 50,

GR mRNA levels increase dramatically, whereas MR mRNA levels decrease. GR

mRNA levels continue to increase towards terrn (approximately 70 days) while

MR mRNA levels remain low (Matthews, 1998). As one can see, GR and MR

receptor systems develop independently in a tirne-specific manner. thus the

outcome of glucocorticoid exposure may differ depending on timing of exposure.

1.6.2 Mechanisms of Autoregulation

Although animal models clearly demonstrate that developing populations

of GR and MR in the hippocampus can be altered, very little is known about the

mechanisrns by which alteration occurs. One possible mechanism is

autoregulation (glucocorticoids acting on GR and MR genes to change their

expression). Autoregulation of GR was detected in in vitro studies that

demonstrated a reduction in GR in cultured cells after prolonged glucocorticoid

exposure (Cidlowski and Cidlowski, 1981; Svec and Rudis, 1981; Bloomfield,

1981). Dexamethasone administration to HeLa cells resulted in decreased GR

protein levels, as established by tritiated glucocorticoid binding. GR mRNA levels

were also decreased, suggesting a decreased rate of transcription (Cidlowski

and Cidlowski, 1981; Svec and Rudis, 1981). Down-regulation of GR rnRNA

levels in HeLa cells was found to be permanent. This down-regulation was

Page 30: Petar - University of Toronto

attributed to a conformational change in the GR promoter (Silva et al, 1994).

Dexamethasone treatment of cells transfected with human GR cDNA produced a

decrease in both GR mRNA and protein. This effect was reported both in

transiently transfected cells (Cos 1, African green monkey kidney cells) and

stably transfected cells (Chinese hamster ovary cells). These studies

demonstrated that human GR cDNA contains sequences critical for homologous

down-reg dation (Bumstein et al, 1 99 1 ).

Elevated concentrations of glucocorticoid within the brain can reduce

corticosteroid receptor expression. The precise mechanism by which such

reduction occurs is not yet known, but several possibilities were postulated

(Bamberger et al., 1996). It is possible that regulation may occur by influencing:

a) intracellular hormone availability, b) interactions between receptor and heat

shock protein, c) receptor phosphorylation, d) nuclear translocation andtor DNA

binding-site modification (kmberger, 1996). lntracellular hormone availability is

regulated by two enzymes, 11 P-hydroxysteroid dehydrogenase (1 1 p-HSD,

isoform 1) and ligand effect modulator 1 (LEMI). II P-HSD metabolizes cortisol

and thus, makes cells cortisol-unresponsive. LEM 1 actively and specifically

exports glucocorticoids from the cell, limiting the glucocorticoid availability (Kralli,

et al. 1995).

Heat shock proteins (hsp) form complexes with glucocorticoid receptorç.

Ligand binding to the complex causes conformational change in the GR, and its

dissociation from hsp molecules. This process can be stirnulated or inhibited by

endogenous regulators, such as a heat-stable protein known as "stimulatof

Page 31: Petar - University of Toronto

(Schmidt, et al. 1985). The stimulator enhances the efficiency of GRlhsp

dissociation. The GRlhsp complex can also be stabilized. The modulator, a

phosphoglyceride molecule, binds to both GR and hsp90, thus stabilizing the

inactive GRJhsp wmplex and therefore negatively regulating GR activation

(Robertson. et al. 1995).

The importance of phosphorylation in the regulation of GR activity is not

well understood.

dissociation from

serine residues (H

GR becomes phosphorylated at multiple sites shortly after

the hsp complex. The phosphorylation occurs primarily on

u, et al. 1994). A possible role of phosphorylation in regulation

of GR activation was derived from studies undertaken in rat fibroblast cells. Such

studies reported increases in GR mediated transcription. during progression from

normal to the neoplastic ceil stage, without the increase in the intracellular GR

levels (Vivanco, et al. 1995). The observed increase in the GR mediated

transcription was attributed to changes in receptor phosphorylztion (Vivanco, et

al. 1995). However, studies undertaken in the mouse, reported that selective

mutations of one, multiple or al1 phosphorylation sites on the GR only modestly

decreased transcriptional activity of the receptor (Mason. et al. 1993). Similar

results were reported for the human GR as well (Almlof, et al. 1995). It is possible

that phosphorylation has a role in intracellular localization of the GR, rather than

its transcriptional activity (Bamberger. et al. 1996).

lntracellular distribution of GR is affected by hsp content and receptor

phosphorylation. Translocation of the GR to the nucleus accelerates after

receptor phosphorylation. It is possible that massive phosphorylation results in

Page 32: Petar - University of Toronto

unmasking of a nuclear signal (Akner, et al. 1994). Hsp molecules are also

actively involved in GR trafficking. Inhibition of hsp56 by FK506 was found to

increase GR-mediated transcription rate by stimulating nuclear transfer of the

receptor (Sanchez, et al. 1994).

Various nuclear factors can influence the binding of activated GR

complexes to DNA. Binding of GR to DNA can occur within specific GRE, other

specific DNA sequences or naked DNA (Bamberger, et al. 1996). Low molecular

radius (700-3000 Da) factor, isolated from rat hepatoma cells and present in

extracts f o m human HeLa cells, is responsible for about 40% of activated GR

binding to calf thymus DNA. in vitro (Cavanaugh, et al. 1994). At present, it is not

known why only some GRs require this factor for binding. Another nuclear factor

involved in GWDNA binding is ATP-stimulated glucocorticoid-receptor

translocation promoter (ASTP). ASTP is a 93kDa histone-binding protein that

increases GR binding to chromatin in the presence of ATP. Helicase proteins,

such as yeast brm proteins or their human homologues, facilitate binding of GR

to nucleosomal DNA (Bamberger, et al. 1996).

Nuclear factors can also have an inhibitory effect on the GR-mediated

transcription. Purification of 'Yranslocationn inhibitor from rat liver demonstrated

the existence of a factor that inhibits binding of activated GR to rat nuclei

(Dahmer, et al. 1985). Similarly, pyridoxal phosphate, the active fom of vitamin

Be, was cismonstrated to inhibit activated GR binding to the DNA. As a result of

this in hibition, GC-induced transactivation decreased by a bout 50% (Allgood, et

al. 1993). Conversely, inhibition of pyridoxal phosphate was found enhance the

Page 33: Petar - University of Toronto

transcriptional effects of glucocorticoids. Cell-specific glucocorticoid sensitivity is

at least partially regulated by pyridoxal phosphate, as levels of this rnolecule Vary

profoundly between different cell types (Allgood. et al. 1993).

At present. the precise involvement of g lucocorticoids in modification of

these mechanisms is unknown. However, it is recognized that glucocorticoids

have the potential to regulate their activity (Bamberger, et al. 1996).

1.6.3 Serotonin and Programming of Corticosteroid Receptors

Serotonin was shown to regulate GR binding levels in primary

hippocarnpal cultures derived frorn fetal rat brain (Mitchell et al, 1990). Incubation

of hippocampal cultures with serotonin increased GR binding to ligand. This

effect was mediated, at least partially, through 5HT2 receptors. Increased GR

binding was found to be long-term as it persisted for 60 days without any

add itional se rot on in exposu re. U nfortu nately, ce1 1-specificity of se rot onin

responses was not addressed in this study. An increase in GR binding has also

been demonstrated in adult rats that were handled as neonates (Anisman et al,

1998; Meaney et al. 1994). Handling exposes the neonates to brief periods of

mild hypothermia. that is associated with thyroid hormone release (Meaney et al,

1994; Mitchell et al, 1990). The increase in GR binding due to neonatal handling

was mimicked by thyroid hormone administration to the neonates (Meaney et al,

1987). Subsequent studies demonstrated that both neonatal handling and thyroid

hormone administration produced an increase in hippocampal serotonin turnover

(Mitchell et al, 1990). As a result. it was proposed that thyroid hormones

Page 34: Petar - University of Toronto

stimulate ascending serotoninergic neurons (originating within the Raphe

nucleus) increasing hippocampal serotonin release and therefore increasing GR

expression (Meaney et al, 1994). Gluwcorticoids were found to influence both

serotonergic systems and thyroid hormone turnover. Prenatal glucocorticoid

exposure is known to increase expression of the serotonin transporter system in

the rat brain (Slotkin et al, 1996) suggesting an advance in the activity of the

serotonergic system. In the fetal guinea-pig. dexamethasone exposure was

found to increase both fetal thyroid hormone and GR mRNA levels (Dean and

Matthews, 1999). Taken together these findings suggest that prenatal

glucocorticoids advance maturation of serotonin system, which could mediate

increases in hippocampal GR mRNA complement.

1.7 Multi Drug Resistance

Studies described in this thesis address the direct mechanisms by which

serotonin and glucocorticoids influence corticosteroid receptors in the developing

hippocampus. Very recently it has been suggested that exposure of the brain to

synthetic glucocorticoids is dependent on the expression of rnulti drug resistance

genes within the brain (De Kloet et al., 1998). To date there is no information as

to whether these proteins are present in the developing brain.

Both the synthetic glucocorticoid dexamethasone and the endogenous

glucocorticoid corticosterone (in rats) were found to be substrates of P-

glycoprotein. P-glycoproteins exclude a large number of seemingly unrelated

substrates from the cytoplasrn and are responsible for the phenornenon of multi-

Page 35: Petar - University of Toronto

drug resistance. Since p-glycoproteins can exclude dexamethasone from the cell

they can protect against actions of dexamethasone. As a result P-glycoproteins

could be involved in the programming of the HPA axis by synthetic

glucocorticoids (De Kloet, et al. 1998). The protective role of 1 P-glycoprotein

against dexamethasone penetration into the brain has been clearly demonstrated

in adult animais (Meijer et al, 1998). Given this evidence and considering the fact

that one of the primary purposes of this thesis was to examine effects of

dexarnethasone on corticosteroid receptor expression, investigation of P-

glycoprotein mRNA expression in the developing brain became a logical

expansion of this study.

The mammalian P-glycoproteins (P-gps) were initially isolated from cancer

cells that were found to be resistant to rnany foms of anti-cancer therapy.

(Sharom, 1997). Closer investigation of anti-cancer therapy resistance led to

discovery of multi drug resistance syndrome (MDR) and identification of P-gps as

primary candidates for mediating MDR. P-gps confer drug resistance through

active. ATP-dependent extrusion of a wide range of cytotoxic drugs. Dnig

extrusion happens against the concentration gradient and is independent of an

electrochemical transmernbrane potential or hydrogen ion gradient (Ruetz et al,

1994). Further investigation of P-gps generated a rather large list of seemingly

unrelated as well as similar substrates (Sharom. 1997). Such findings

characterized P-gps as unusual transporters whose actions can be both harmful,

as in MDR, and beneficial, as in their protective role against penetration by many

xenobiotic compounds. Human P-gps are encoded by two distinct genes:

Page 36: Petar - University of Toronto

MDR1 and MDR3. Rodent P-gps (at least in mouse in rat, other rodent species

are poorly described) are enwded by three distinct genes: mdrla. rndrl b and

mdR. Human MDRI , and mouse mdrl a and mdrl b genes confer multi drug

resistance. These genes are often referred to as mdrl-type P-gps or class I .

Human MDR3 and mouse mdR do not confer multi drug resistance (Chen et al,

1986; Gross et al, 1988).

1.7.1 Structure and Substrates

P-glycoprotein is a 170 kDa plasma membrane glycoprotein. It belongs to

the ABC superfamily of proteins. which are ATP-dependent transporters.

Structural analysis of P-gps revealed the presence of two membrane-bound

domains, each consisting of six trans-membrane segments (a-helices), and two

ATP cytosolic domains (Sharom 1997). Domains are arranged in toroidai

configuration, when viewed from a bove the membrane, with six-fold symmetry.

Pgp has a diameter of -1Onm with a central pore -5nm in diameter, as reported

by electron microscopy study (Rosenberg et al, 1997). Twelve transmembrane

segments are arranged in two symmetncal halves, with helices six and twelve

being in close proximity (Loo et al, 1996). P-glycoprotein has a large number of

substrates. The largest group of MDR substrates are the numerous anti-cancer

drugs. These include Vhca alkaloids, taxanes, anthracyclines and

epipodophylllotoxins (Sharom 1997). Another large group of MDR substrates

includes other cytotoxic agents, linear and cyclic peptides and certain steroids

(Sharom 1 997). Typical MDR substrates are molecules with molecular weight

Page 37: Petar - University of Toronto

greater than 400KDa. that are highly hydrophobic and amphipathic. Typical

substrates usually have planar ring system and often cany a positive charge at

physiological pH. However, many other MDR su bstrates differ from this

description, as they are found to be hydrophilic, linear and uncharged at

physiological pH (Eythan et al, 1994; Loe et al, 1994; Sarkadi et al, 1994;

Sharma et al, 1992; Sharom 1995; Sharom et al, 1996).

1.7.2 Physiological Role of P-gp

A physiological role of P-gp has been pursued since its discovery. Several

studies indicated a function in transport of lipids and steroids. Mouse Pgp class

II, as well as hurnan Pgp class I are highly expressed in the adrenal gland.

Mouse P-gp class I I (also designated mdrl b) is also hig hly expressed in the

pregnant uterus. Human P-gp is found in human placental trophoblasts and

secretory endometrium (Thiebaut et al, 1987; Croop et al, 1989). Such high

expression of Pgp in reproductive and hormone producing organs suggested its

role in reproduction and hormone production. This idea was also supported by

the finding that Pgp could transport hormones cortisol, corticosterone and

aldosterone (Ueda et al, 1992; Bourgeois et al, 1993). On basis of such findings

it was proposed that P-gp wuld play a role in rapid secretion of hormones or

even in protection of hormone producing cells against the high intracelluiar

hormone accumulation. However, such speculations remain controversial.

The greatest insight into the physiological role of P-gp was offered with

creation of MDRla knockout mouse, as well as mouse deficient for al1 of the

Page 38: Petar - University of Toronto

MDR genes. Examination of the MDR knockout mouse (-Io) revealed that they

differ from the wild type only in their sensitivity to xenobiotic compounds,

especially those that target central nervous system (Schinkel et al, 1997). All

other aspects of physiological function were found to be identical (Schinkel et al,

1997). It was established that P-gp class I (mdrla) is not essential to basic liver,

kidney or intestinal function, as well as to the functions of the brain, adrenal

gland, ovaries and pregnant uterus. In addition, hematopoietic stem cells ar;d the

tiematological compartment in general are also unaffected by absence of P-gp

class 1. Taken together these findings suggest that function of P-gp class I is

either taken over by some related transporter or that it simply plays no significant

role in these physiological functions (Schinkel et al, 1997).

MDR knockout mice show greatly enhanced susceptibility to a range of

xenobiotic compounds such as: ivermectin. vinblastine, digoxin, cyclosporin A,

loperamide, domperidone and ondanestron (Schinkel et al, 1996). lncreased

penetration of these compounds into the central nervous systern is pronounced.

Exposure of MDR knockouts to ivermectin, a neurotoxic pesticide is lethal.

lverrnectin levels inside the brain of knockout mice were found to be 100 times

higher when compared to wild type (Schinkel, 1995). Sirnilariy, a subpopulation

of CF-1 mice, that do not express mdrla, showed 100% susceptibility to cleft

palate syndrome, when exposed to L-652,280 (the 8,9 Z photoisorner of the

naturally occumng avermectin 1 B) (Lankas et al, 1998). This cornpound is known

to induce cleft palate formation. Heterozygous mice, P-gp (+/-), showed

decreased susceptibility and wild type mice. which express mdrla fully, were

Page 39: Petar - University of Toronto

found to be completely insensitive to L-652,280, at the doses tested. The same

study discovered that the degree of chernical exposure of fetuses was inversely

related to the expression of placenta1 P-gp. Such a finding cleariy dernonstrated

the protecüve role of rndrla against potential teratogens (Lankas et al, 1998).

Dexamethasone, a synthetic glucocorticoid used clinically, is also a substrate for

P-gp. It's penetration into the brain of knockout rnice is three fold higher then in

wild type. P-gp could hence offer protection against actions of dexamethasone in

the developing brain (Schinkel et al, 1995).

1.7.3 Expression of MDR

Expression of each MDR gene is highly tissue specific. Mdrl a is highly

expressed in the blood-brain barrier, blood-testes barrier, placenta and intestinal

epithelium (Nakamura et al, 1 997). Mdrl b is highly expressed in the adrenal

gland, pregnant uterus. and ovaries. 80th genes are also expressed in an

overiapping fashion in the liver, kidney, lung and heart tissue

Schinkel et al, 1994).

Expression of MDR has also been examined in

(Croop et al, 1989;

the human fetus.

Expression of MDRI mRNA was observed as eariy as embryonic week 7 (Van

Kalken, 1992). The definitive zone of fetal adrenal gland showed expression of

MDR1 which increased throughout gestation. There was no detectable

expression of MDRl wlhin the fetal zone of fetal adrenal gland. Fetal intestine

showed no expression in eariy gestation, but expression was increased in late

pregnancy. Expression was also detected in main bronchi and pharynx, which do

Page 40: Petar - University of Toronto

not express MDRI in adults. Expression of MDRI within kidney and liver was

detectable at eleven weeks of gestation. Expression of MDRl within the brain

capillaries was not observable before the third trimester (Van Kalken. 1992).

Recent studies indicated that expression of MDR could be induced or

suppressed by its substrates. Exposure of Caco-2 cells to verapamil, celiprolol

and vinblastine increased P-gp expression when compared to control. In

contrast, exposure to metkephamid decreased P-gp expression when compared

to control (Andrele et al, 1998). This finding is certainly in agreement with the

development of MDR in tumor cells after exposure to cytotoxins. It would be

interesting to see whether P-gp induction can be observed in normal tissues as

well, after exposure to a substrate.

Page 41: Petar - University of Toronto

Chapter 2: Rationale and Objectives

2.1 Clinical Importance

Up to 10% of pregnant women in North America, at risk of preten

delivery. are treated with synthetic glucocorticoids. This is done to prornote fetal

organ maturation (NI H, 1 995). Synthetic glucocorticoid treatment is often

implemented in early gestation and in some cases throughout gestation. At

present, very little is known about the impact of such treatment on brain and

neuroendocrine developrnent (Matthews. 2000).

Epidemiological studies in humans have associated an adverse

intrauterine environment with subsequent development of coronary heart

disease, hypertension, hyperlipidemia and non-insulin dependent diabetes

mellitus (reviewed in Baker, 1991). In addition, sustained fetal exposure to

elevated levels of endogenous glucocorticoids was associated with low birth

weight, developmental delays and neurological disorders (Field, et al. 1985;

Ward. 1991 ; Clernent, et al. 1992). Emerging evidence indicates that alteration in

development of the fetal HPA axis may represent a link between fetal

environment and the development of the adult disease.

2.2 Rationale

Animal studies demonstrated that increased fetal exposure to

glucocorticoids can lead to permanent alteration in HPA function. The alteration

in HPA function was associated, to a great extent, with changes in hippocampal

Page 42: Petar - University of Toronto

corticosteroid receptor expression (Levitt, et al. 1996; Dodic, et al. 1 997; Dean, et

al. 2000; Matthews, et al. 2000). The alteration in HPA function was linked to

development of hypertension in adult animal (Levitt, et al. 1996). At present,

there is little information on the direct effects of glucocorticoid exposure on

expression of corticosteroid receptors in fetal hippocam pal neurons. The main

purpose of this study was to examine this question.

Expression of multi drug resistance genes in adult animals was found to

protect against dexamethasone exposure (Meijer, et al. 1997). It has been

suggested that the expression of multi drug resistance genes in fetuses could

have the same effect. Since multi drug resistance gene expression limits cellular

exposure to dexamethasone it probably diminishes the ability of dexamethasone

to change corticosteroid receptor expression. To date there is no information as

to whether these genes are expressed in the developing brain. Therefore,

examination of the multi drug resistance gene expression in the developing brain

was a necessary step in an atternpt to describe the role of muiti drug resistance

genes in the HPA programrning.

2.2 Objectives

The first objective in the mouse study was to set up a mouse hippocampal

culture system that would allow efficient in situ hybridization analysis of GR and

MR mRNA expression levels. The second objective was to examine direct effects

of dexamethasone, corticosterone and serotonin on the expression of GR and

MR mRNA in cultured fetal hippocampal neurons, in vitro (Chapter 3).

Page 43: Petar - University of Toronto

Examination of this question in the mouse was necessary in order to compliment

the existing evidence of HPA programming, in species with a postnatal profile of

neuroendocrine developrnent (in species where maturation of neuroendocrine

systems occurs primarily after the birth).

The primary objective of this thesis was to examine direct effects of

dexamethasone, cortisol and serotonin on the expression of GR and MR mRNA

in fetal guinea-pig neurons, in vitro (Chapters 4 and 5). Guinea-pigs have a

prenatal profile of neuroendocrine development (in this species maturation of

neuroendocrine systems occurs primarily prior to birth) and thus, resemble the

human more closely than rodent species such as the rat and mouse.

Examination of GR and MR mRNA expression was carried out at different stages

of developrnent.

The primary objective of the multi drug resistance study (Chapter 6) was

to examine the expression of MDRla gene in the guinea-pig brain throughout

development. The second objective of this study was to determine whether brief

periods (2 days) of dexamethasone exposure in fetal life alters the expression of

the MDRl a gene.

Page 44: Petar - University of Toronto

Chapter 3

Cel l Specific Regulation and mRNA

Expression in Mouse Hippocampal Culture

3.1 Introduction

The hippocampus plays a central role in regulation of the hypothalamo-

pituitary-adrenal (HPA) axis (Heman, et al. 1996; Jacobson, et al. 1991). High

levels of glucocorticoid receptor (GR) and mineralocorticoid receptor (MR) are

present within the hippocampus and extensive evidence indicates that

glucocortwid feedback of the HPA axis occurs at this level (De Kloet, et al.

1998).

It is well established that HPA function can be programmed during

development (Levine, et al. 1957). Such programming occurs, at least partially, at

the level of the hippocampus (Levine, et al. 1957; Bakker, et al. 1995; Liu, et al.

1997; Plotsky, et al. 1993; Matthews, 2000). Administration of synthetic

glucocortiwid (dexamethasone) to pregnant rats results in adult offspnng that

exhibit elevated basal plasma corticosterone concentrations. This has been

associated with reduced levels of GR and MR mRNA in the hippocampus (Levitt,

et al. 1996). The mechanism and time course by which GR and MR complement

are pemanently altered remains unknown. Recent studies in the guinea pig

indicate that prenatal dexamethasone exposure has acute effects on

hippocampal GR and MR mRNA levels, in fetuses and neonates (Dean, et al.

Page 45: Petar - University of Toronto

1999). However, it is unclear whether glucocorticoids act directly at the

hippocampal receptor or at other sites in the brain to influence GR and MR

Ievels.

Serotonin is known to be involved in maturation and regulation of the HPA

axis (Dinan, 1996). It is well established that serotonin can regulate GR

expression and binding, both in vivo and in vitro (Mitchell, et al. 1990; Mitchel, et

al. 1990). Administration of serotonin to rat hippocampal neuronç, in vitro, causes

long-ten increases in GR expression as well as binding (Mitchel, et al. 1990).

However, it remains unclear whether this change occurs in neurons andlor glia.

3.2 Objectives

In this study, we investigated the direct effects of endogenous

glucocorticoids (corticosterone) and synthetic glucocorticoids (dexamethasone)

as well as serotonin on GR and MR mRNA levels within mouse hippocampal

neurons and glia, in vitro. We utilized a technique that allowed us to examine GR

mRNA and MR mRNA expression in a cell-specific manner.

3.3 Hypothesis

We hypothesize that exposure to dexamethasone will down regulate GR

mRNA expression in fetal mouse hippocampal neurons. MR mRNA expression

will not be affected by the dexamethasone exposure.

Corticosterone treatment will down regulate both GR and MR mRNA

expression in fetal mouse hippocampal neurons.

Page 46: Petar - University of Toronto

Exposure to serotonin will up regulate GR mRNA expression in

hippocampal neurons. MR mRNA expression will not be affected by exposure to

serotonin.

These speculations are based on the fact that dexamethasone binds

specifically to the GR with minimal biniding to the MR. In contrast, corticosterone

binds to both GR and MR. and thus, has the potential to influence expression of

both GR and MR mRNA. Effects of serotanin on the expression of GR and MR

mRNA are based on previous findings in related studies.

3.4 Methods

3.4.1 Mouse hippocampal culture

Accurately time-dated pregnant CD1 mice were obtained from

Charles River Laboratories (St-Constant, Quebec, Canada). All animal

experiments were carried out according to protocols approved by the Animal

Care Cornmittee at the University of Toronto, in accordance with the Canadian

Council for Animal Care. Animals were killed on day 18 of pregnancy by crevical

dislocation. Dead animals were placed on a paper towel previously soaked in

ethanol (70%) and sprayed with ethanol (70%). An abdominal incision was made

and skin removed. Another incision was made to the underlying connective

tissue and muscle tissue in order to allow removal of fetuses. Care was taken not

to puncture intestinal organs.

Fetuses were placed into a petri dish (60mm) and transfened to a tissue

culture hood. Fetal membranes were dissected and fetuses removed to fresh

Page 47: Petar - University of Toronto

petri dish (60mrn) containing dissection medium (see Appendix 1). Fetuses were

decapitated and fetal skin and dural membranes were removed. Fetal skulls were

cut along the sagital suture and brains were isolated and placed into a fresh petri

dish (60mm). containing dissection medium. Under a dissecting microscope, fetal

brains were separated into hemispheres. This was done without cutting, by

pushing the hemispheres apart. Hippocampi from each hemisphere were

removed using pointed forceps and a scalpel blade (No.10). All hippocampi were

transferred to a clean petri dish (35mm) containing dissection medium.

Dissection medium was replaced with 1.5mI trypsin\EDTA (see Append ix

1). The hippocampi were incubated for 15 min at 37°C. The trypsinlEDTA

solution was removed and hippocampi were placed in a 15ml falcon tube

containing 1.5ml of MEM - HS, FBS (see Appendix 1). Hippocampi were further

dispersed by trituration in MEM-HS, FBS. Hippocampi were passed through a

pasteur pipette (10 times) and then through a flame-polished pasteur pipette (10

times). Dissociated hippocampal cells were suspended in MEM-HS, FBS (6 ml).

The cell suspension (50pL) was placed in each well containing MEM-HS, FBS

(250pL). A total of 8 wells per culture slide were prepared. 6 culture slides per

animal. The day of dissection was counted as day O (DO). On DO + 4 medium

was changed in each well. FUDR (20& 0.02M) was added to each well when

glial cells within the wells were found to be confluent. FUDR addition was

necessary in order to prevent glial cell overgrowth.

On DO+iO treatment was initiated. Treatment groups included 5HT,

corticosterone and dexamethasone. Each treatment was adrninistered at 1 nM,

Page 48: Petar - University of Toronto

1 OnM and 1 OOnM concentrations, respectively (see Appendix 2). Each slide had

two control wells, 2 X IOOnM wells, 2 X 10nM wells and 2 X InM wells, for each

treatment respectively. Each treatment was carried out in duplicate and at least

six different experiments were carried out per treatment. Treatment was repeated

for four days. at 24-hour intervals. On D0+14 cultures were fixed with 4%

parafomaldehyde (see Appendix 3) and stored under 95% ethanol, at 4"C, until

processing with in situ hybridization.

3.4.2 IN SITU HYBRlDlZATlON

All solutions used in this section were described in detail in Appendix 3.

3.4.3 Oligonucleotide Probes

The GR oligonucleotide probe used in rnouse cultures was complimentary

to bases 1021-1053 of the coding sequence of mouse GR mRNA (Danielsen et

al, 1986). The MR oligonucleotide probe used was complimentary to bases 2942-

2986 of human MR mRNA (Ariza et al, 1987). The anti-sense GR and MR

oligonucleotide probes were synthesized using an Applied Biosystems DNA

synthesizer (mode1 392).

3.4.4 Probe Labeling

Tailing buffer (5X, 2pL, Gibco BRL) was added to each reaction tube. The

oligonucleotide probe (1 PL, 1 UnglpL) was added directly to tailing buffer. 3 5 ~

deoxyadenosine-5'-a-thiotriphosphate (APL, 1300 Cümmol, NEN, Du Pont

Canada Inc.) was added to the reaction mixture. 35~-incorporation into

Page 49: Petar - University of Toronto

oligonucleotide probe was achieved with addition of terminal dioxynucleotydyl

transferase (1.5pL, Gibco BRL). Finally, DEPC-treated water (14pL) was added

to the mixture. Reagents were rnixed by gentle passage up and down the pipette.

The mixture was incubated in water bath for 1 h, at 37°C. EDTA (2pL, 0.5M) and

STE (28pL, 1.OM) buffers were added to stop the reaction. The separation of

bound and free probe was achieved by passing the reaction mixture through

sepadex spin columns ( ~ r o b e ~ u a n t ~ ~ G-50 Micro Columns, Amersham

Phamacia Biotech Inc.), which were centrifuged for 3min at 3000rpm.

Dithiolthreitol (2pL) was added to labeled probe to prevent disulfide bond

formation and probe degradation. The radioactivity of the final mixture was

measured using a p-scintillation counter.

3.4.5 Hybridization

Prior to application of labeled probes the plastic charnber sections and

silicone gaskets were removed from the giass slides, which-were allowed to dry

completely. Radio-labeled probe was added to hybridization buffer (Appendixd)

so that final activity in the buffer was 1200 cprn/pL. Each culture-slide was

covered with hybridization buffer (1 80pL). Culture-slides were then covered with

çteriie parafilm. Care was taken to expel air bubbles undemeath the parafilm.

Moisture levels in the incubation chamber were maintained with soak solution

(Appendix3). Slides were incubated overnight at 42OC.

Antisense probes were also incubated with rnouse brain cryo-sections (10

pm). Frozen mouse brain was coronally sectioned (12pm) using a cryostat

Page 50: Petar - University of Toronto

(JUNG CM3000, Leica instruments GrnbH, Nussloch. Germany). Sections were

thaw-mounted on poly-L-lysine wated glass microscopie slides. Sections were

stored at -80°C. Prior to hybridization the sections were fixed in 4%

parafomaldehyde solution (5rnin). Slides were washed in phosphate buffer

saline (PBS) (Wlmin) and dehydrated in 70% ethanol (5min) and 95% ethanol

(1 min).

3.4.6 Washing

Culture-slides were placed in 1XSSC solution and the parafilm was gently

rernoved. Slides were washed in lXSSC (room temperature, for 20min) and then

in 1XSSC for 30min at 55OC. Finally, slides were rinsed in IXSSC, 0.1 %XSSC,

ethanol (70%) and ethanol (95%), at room temperature.

3.4.7 Emulsions

Emulsion autoradiography was camed out in the dark room.

Radiosensitive photographic emulsion (Ilford Scientific, K.5) was allowed to rnelt

prior to use (42OC water bath). Emulsion solution was diluted to 50% with

ultrapure (millipore) water. Emulsion was placed in specially designed glass

beaker which aIlowed emulsion thickness to be constant across the slides.

Ernulsion-coated slides were allowed to dry in the dark for 1 h prior to their

storage at 4OC. Exposure time for MR mRNA was 8 weeks and for GR mRNA

was 10 weeks.

Page 51: Petar - University of Toronto

3.4.8 Developing and staining emulsions

Emulsion coated slides were placed in developer (Kodak D520) for 5 min

and then transferred into stop bath (Kodak) for 1 minute. Finally, slides were

moved into liquid fixative (Kodak) bath for a further 5 min. Slides were then

rinsed extensively in ultra water prier to counter staining.

Developed ernulsion-coated slides were counter stained with methylene blue

(0.01%). Methylene blue is a positively charged (cationic) dye which combines

strongly with negatively charged cellular constituents such as nucleic acids and

acidic polysaccharides. It is an excellent general dye used to stain cells and

increase their contrast so that they can be more easily seen in the bright-field

microscope (Brock, et al. 1994). Slides were processed in the following order:

PBS (2min), methylene blue (OBI%, made in PBS, 10-20s), ultrapure water

(20s), ethanol (50%, 2min), ethanol (70%, Gmin), ethanol (go%, 7min) and xylene

(3min). After xylene exposure each slide was mounted with mounting compound

(Tissue Tek). Slides were allowed to dry ovemight.

3.4.9 Cell Viability

Viability of cells in culture was determined by trypan blue staining. Mouse

cultures were prepared and treated as previously described. Treatment included

only the highest doses (100nM). After the treatment, trypan blue (04%, 20pL)

was added to each well of a culture-slide. Culture-slides were allowed to stand

for 2 minutes prior to counting.

Page 52: Petar - University of Toronto

3.5 Data analysis

Autoradiography revealed high levels of GR and MR mRNA in the

hippocampus as well as GR mRNA distributed in the areas of the brain (Fig.3.1).

This is consistent with the previously reported distribution of these receptors

(Matthews, 1998; Levitt, et al. 1996). A sense probe dernonstrated no

hybridization in an adjacent section known to contain GR and MR mRNA

(Fig.3.1).

Analysis of trypan blue stained cells was camed out at 50X magnification.

On average, up to 100 cells per well were examined for presence of trypan blue.

In total, less than 5% of cells were identified as trypan blue positive. To confirm

the validity of trypan blue test, cells were allowed to stand under the microscope

after the initial count was done. After approximately 10min drastic increase in the

number of trypan blue positive cells was observed, thus confirming the validity of

the test.

The laboratory has extensive experience in using liquid emulsion

autoradiography and great care was taken to ensure that emulsion thickness was

consistent across the slides. Care was also taken to avoid saturation of silver

emulsion, which leads to non-linearity of analysis. Analysis of GR mRNA and MR

mRNA in pyramidal neurons was camed out at 200X rnagnification. On average

30-80 grains were identified over positive neurons. Mean grain count per neuron

was calculated for each treatment (6 separate experiments per treatment).

Approximately 15 mRNA positive neurons were randornly sampled and analyzed

in each well (2 wellsltreatment/analysis). Positive neurons were selected on

Page 53: Petar - University of Toronto

basis of MR mRNA expression and cellular morphology. Within the CNS, MR

mRNA is expressed only in neurons (De Kloet et al., 1998). In addition selected

cells were found to have typical neuron cell morphology, such as the large

nucleus, large and visible nucleolus and multiple processes (Banker and Goslin,

1991). MR mRNA expressing cells were also found to retain more methylene

blue stain. In case of neurons that were analyzed for GR mRNA expression,

selection was based only on cellular morphology and methylene blue staining.

Random sampling was achieved by selecting neurons for analysis at 50X

magnification. At this magnification level silver emulsion grain expression

between neurons is not distinguishable, and thus selection is blind with respect to

silver emulsion grain expression. Grain counting was also performed in a blinded

fashion, with respect to treatment group. AH neurons within the view field were

included in analysis, even if grain expression in specific neuron was not detected.

Cell counting revealed approximately 100 neurons/well. Grain counting over

regions of confluent glial cells was undertaken using a cornputen'zed image

analysis system (Imaging Research, St. Catherines, Ontario, Canada)(Matthews,

et al. 1995; Matthews and Challis, 1995). Group data are presented as

MeansS.E.M. Statistical analysis was performed using Statistica (Release 5, 97

Edition, Oklahoma, USA). The effects of treatment on GR and MR mRNA were

determined using a one-way ANOVA followed by the Duncan's method of post-

hoc comparkon. Significance was set at pe0.05.

Page 54: Petar - University of Toronto

3.6 Results

Glucocorticoid receptor mRNA levels were high in the hippocarnpal

pyramidal neurons as well as in glia. High levels of mineralocortirnid receptor

mRNA were detected in the pyramidal neurons. with no expression in glial cells.

This distribution was consistent with that obsewed in whole brain sections

following hybridization with the same probes (Fig.3.la and b).

Dexamethasone exposure (1 0nM and 100nM) significantly ( ~ ~ 0 . 0 5 ) down-

regulated GR mRNA levels within neurons (Fig.3.2a). while GR mRNA levels

were no different between dexamethasone treatment (1 nM) and control wells. In

wntrast, dexamethasone treatment (100nM) had no significant effect on MR

mRNA levels (Fig.3.2b).

Treatment of hippocampal cultures with corticosterone at either 100nM or

1 OnM significantly (p<0.015) down-regulated GR mRNA expression (Fig.3.3a).

Exposure to corticosterone (InM) did not significantly alter GR mRNA levels.

Corticosterone administration (1 00nM) had no significant impact on MR mRNA

levels (fig.3.3b).

Serotonin exposure (1 00nM) resulted in significant (pc0 .Os) u p-regulation

of GR mRNA levels within neurons, where as exposure at lOnM and InM was

without effect (Fig .3.4a). MR mRNA expression was u naffected by serotonin

exposure (100nM) (Fig.3.4b). Analysis of glial cells revealed no significant effect

of any treatment (dexamethasone. corticosterone, serotonin) on GR mRNA

levels (Table 1 ).

Page 55: Petar - University of Toronto

Exposure to any of the treatments was found to have no effect on

proportion of GWMR mRNA positive to negative cells. Analysis of al1 treatments

revealed that more than 95% of examined cells were G W M R mRNA positive.

3.7 Discussion

In the present study we have demonstrated, for the first time, that

glucocorticoid exposure modifies GR mRNA levels in a cell-specific manner. Both

synt hetic glucocorticoid (dexamethasone) and endogenou s glucocorticoid

(corticosterone) decreased GR mRNA levels in hippocampal neurons but not

glial cells. We have also identified that se rot onin exposure (1 OOnM) increases

GR mRNA levels in neurons but not glial cells.

It has been previously demonstrated that fetal exposure to glucocorticoids

leads to long-term changes in HPA activity in rats, guinea pigs and primates

(Bakker, et al. 1995; Levitt, et al. 1996; Dean, et al. 1999; Muneoka, et al. 1997;

Uno, et al. 1990). Exposure of fetal rats to glucocorticoids over the last week of

gestation results in adult offspring with increased basal adrenocortical activity

and this was associated with increased blood pressure (Levitt, et al. 1996).

Analysis of prepubertal rat offspring following exposure to glucocorticoids on day

l7, l8 and 19 of pregnancy revealed increased corticosterone responses to

stress (Muneoka, et al. 1997). Elegant studies in the rat have demonstrated that

increased basal corticosterone in adult offspring, that had previously besn

exposed to glucocorticoids in utero, is associated with a reduction in

hippocampal GR and MR mRNA levels (Levitt, et al. 1996; Dean, et al. 1999). No

Page 56: Petar - University of Toronto

differences in corticosteroid receptor mRNA were noted in other brain regions.

Together these data indicate that the 'programmed' increase in HPA activity is

linked to a reduction in glucocorticoid feedback sensitivity. Results from the

present study would suggest that there are acute effects of synthetic

glucocorticoids on GR production in fetal hippocampal neurons but no direct

effect on MR populations. However, the relatively limited life span of mouse fetal

neurons in culture (approximately 21 days) makes it difficult to establish whether

the acute direct effects of glucocorticoids on GR synthetic processes are

permanent.

Changes in GR mRNA levels following synthetic glucocorticoid exposure

have been reporteci in other in vitro culture systems (Cidlowski, et al. 1 981 ; Svec,

and Rudis. 1981; Webster and Cidlowski. 1994). One such study demonstrated

that dexamethasone can perrnanently down-regulate expression of GR mRNA in

HeLa cells. This permanent down-regulation was associated with modification

(methylation) in the GR promoter (Silva, et al. 1994). It is possible that

glucocorticoids induce similar changes to the GR promotor in developing

neurons, though this rernains to be detenined.

The failure of glucocorticoids to modify MR mRNA levels in the present

study would impiy that the effects of glucocorticoid exposure on MR mRNA levels

demonstrated in adult rat offspring (Levitt, et al. 1996) occur through an indirect

mechanism. Dexamethasone binds mainly to GR (Kd-5nM) and has very low

afinity for the MR (Kd-5OnM) (Veldhius, et al. 1982). Therefore, it is perhaps not

surprising that dexamethasone was found to have direct effects on GR mRNA

Page 57: Petar - University of Toronto

levels but no effect on MR mRNA (at 1 and IOnM concentrations). However, it is

possible that dexamethasone activated MRs at 1 OOnM concentration. Absence of

effect on MR mRNA expression at IOOnM concentration irnplies that

dexamethasone dose not regulate MR mRNA expression directly. Corticosterone

has 10-fold higher affinity towards MR than GR (Rupprecht, et al. 1993; Anna,

etal. 1988; Evans and Ariza. 1989; Cato, et al. 1991). Hence, the absence of

any significant direct effect of corticosterone or dexamethasone exposure on MR

mRNA supports the suggestion that glucocorticoids exert their influence on MR

synthetic processes via indirect pathways.

It is accepted that prenatal programming of the HPA axis occurs, to a

large part, at the level of hippocampal GR and MR (Matthews, 2000). Both the

hippocampal GR and MR act to suppress HPA activity (Jacobson and Sapolsky,

1 991 ). Evidence indicates that the MR is involved in feed back regulation of basal

HPA function while the GR is important during periods of HPA activation (De

Kloet et al., 1998). If similar GR autoregulation, to that which we report in vitro,

occurs in vivo, then this may itself have very significant consequences for normal

hippocampal development. Longitudinal studies examining acute as well as long-

term effects of fetal glucocorticoid exposure are clearly warranted.

A role of serotonin in the regulation of GR development in primary

hippocampal cultures, derived frorn fetal rat brain, has previously been

described. Meaney and colleagues demonstrated that 4 days of serotonin

exposure was sufficient to pemanently increase GR binding. It has subsequently

been esta blis hed that this effect is mediated via high-affinity 5HT2 receptors

Page 58: Petar - University of Toronto

(Kd-5nM) (Mitchel, et al. 1990). However, it has remained unclear whether the

increase in GR levels occurred in neurons, glia or both. In the present study, we

clearly demonstrate that increases in GR mRNA following serotonin exposure are

neuron-specific. A possible functional limitation in this part of the study was the

low stability of 5HT in solution, as 5HT is known to oxidize within one to two

hours in solution (Mitchel et al., 1990). In the future this limitation can be

overcorne by use of specific 5HT2 receptor agonists, which are much more stable

in solution. Use of 5HT2 receptor agonists/antagonist in a sirnilar study would

also confirm the specificity of 5HT receptor involved in mediation of the response.

Meaney and colleag ues also elegantl y demonstrated that central

serotonergic systems were responsible for the increase in GR binding observed

in rats that were handled as neonates. Neonatal handling is associated with

thyroid hormone release (Mitchell. et al. 1990; Meaney, et al. 1994) and thyroid

hormone administration to neonatal rats increased hippocampal serotonin

turnover (Meaney, et al. 1987). However, thyroid hormone administration had no

effect on hippocampal GR or MR mRNA levels, in vitro (Meaney, et al. 1994). It

has subsequently been proposed that a thyroid-serotonin pathway plays an

integral role in regulating GR levels during development. Dexamethasone has

been found to promote maturation of serotonergic systems (Slotkin, et al. 1996),

and in the guinea pig, dexamethasone administration was found to induce an

acute increase in fetal thyroid hormone, as well as hippocampal GR mRNA levels

(Dean, et al. 1999). Based on these findings one can conclude that serotonin

plays an important role in the programming of hippocampal corticosteroid

Page 59: Petar - University of Toronto

receptors. Our findings certainly support such a view, and indicate a direct effect

of serotonin on hippocampal pyramidal neurons. Therefore, it appears that

glucocorticoids can act both to suppress and increase GR mRNA expression. By

acting directly on hippocarnpal pyramidal neurons glucocorticoids down-regulate

GR mRNA expression, as demonstrated by the present study. However,

glucocorticoids also have the potential to indirectly increase GR expression

within hippocampal pyramidal neurons through actions on thyroid hormone and

on maturation of the serotonergic system (Dean, et al. 1999; Slotkin, et al. 1996).

Therefore, it is likely that the outcome following glucocorticoid treatment depends

on the developmental stage of the central newous system at the time of

exposure. If serotonergic systems are immature at the time of glucocorticoid

exposure, one would predict a decrease in hippocampal GR complement.

However, if serotonergic systems are mature at the time of glucocorticoid

exposure, the autoregulatory effect (decrease in GR mRNA) may be ovemdden

by serotonergic ddve, resulting in a net increase in GR rnRNA complement.

In summary, our results indicate that exposure to glucocorticoids has a

direct cell-specific effect on GR mRNA expression in hippocampal neurons.

Furthenore, we have shown that exposure to serotonin increases the GR

mRNA complement within hippocampal neurons. We speculate that both

glucocorticoids and serotonin exert their influence on the developing

hippocampus, but that the net effect of such treatme~t is dependent on the

maturational stage of both systems, at the tirne of exposure.

Page 60: Petar - University of Toronto

Figure 3.1: A) Digitized image of GR mRNA and MR mRNA (8) in coronal sections of an adult

mouse brain following in situ hybridization. C) Fetal hippocampal neurons in culture. D)

Autoradiographic image of a coronal section incubated with a sense probe.

Page 61: Petar - University of Toronto
Page 62: Petar - University of Toronto

Figure 3.l b: Silver emulsion grains over rnouse hippocampal neurons. Black dots represent clusters of GR mRNA.

Page 63: Petar - University of Toronto
Page 64: Petar - University of Toronto

Figure 3.2

A) Analysis of GR mRNA levels following dexamethasone (0, 1. 10, 100nM) exposure (4 days, n=8). Results are expressed as meantS.E.M. *Indicates statistical difference (pc0.01) from control.

B) Analysis of MR mRNA levels following dexamethasone (O, 1, I O , 100nM) exposure (4 days. n=8). Results are expressed as m e a n ~ S .E.M.

Page 65: Petar - University of Toronto

Figure 3.2

O 1 10 100

Treatments (nM)

Treatments (nM)

Page 66: Petar - University of Toronto

Figure 3.3

A) Analysis of GR mRNA levels following corticosterone (0. 1 , 1 0, 100nM) exposure (4 days, n=8). Results are expressed as meanf S E M . 'Indicates statistical difference (pcO.01) from control.

B) Analysis of MR mRNA levels following corticosterone (0. 1, 10, 100nM) exposure (4 days, n=8). Results are expressed as meanfS.E.M.

Page 67: Petar - University of Toronto

Figure 3.3

O 1 10 1 O0

Treatment (nM)

0 1 O0

Treatrnent (nM)

Page 68: Petar - University of Toronto

Figure 3.4

A) Analysis of GR mRNA levels following serotonin (O, 1, 10, 100nM) exposure (4 days. n=8). Result: are expressed as rnean+S.E.M. *lndicates statistical difference (pc0.01) from control.

B) Analysis of MR mRNA levels following serotonin (O, 1, 10, 100nM) exposure (4 days, n=8). Result: are expressed as rnean2S.E.M.

Page 69: Petar - University of Toronto

Figure 3.4

O 10 1 O0

Treatment (nM)

Treatment (nM)

Page 70: Petar - University of Toronto

Table 1

Table 1. Analysis of GR mRNA levels in glial cells. following dexamethasone,

corticosterone and serotonin treatment (n=8). Dexamethasone, corticosterone

and serotonin were administered at 1 OOnM, 10nM and 1 nM concentrations.

Controls (OnM) received vehicle alone.

Treatments (n=8)

Corticosterone

Results are expressed as meankS.E.M number of silver emulsion grains per

4x1 oJ mm2.

Dexamethasone Serotonin

Treatment concentrations OnM

275.2k5.4 201.3k29.0 229.6k11.4

1 nM 250.019.7 192.7k32.1 228.7+1 0.6

10nM 255.0I8.6 189.3Q6.7 228.4t11.2

100nM 256.9k9.7 191.7k27.7 232.1 k7.4

Page 71: Petar - University of Toronto

Chapter 4

Regulation of GR and MR mRNA During Development:

Guinea-pig Studies, Gestationai Age 40 Days

4.1 Introduction

Long-terni modifications in hippocampal GR and MR levels have been

associated with permanent alteration of basal and stress-induced HPA function

(Bakker, et al. 1995; Jacobson. et al. 1991; Levitt, et al. 1996; Liu, et al. 1997;

Plotsky, et al. 1993). Studies undertaken in rats have dernonstrated that fetal

exposure to synthetic glucocorticoids can lead to offspring that express

decreased levels of GR and MR mRNA in the hippocampus and dentate gyrus.

These offspring also had elevated basal plasma corticosterone concentrations

and were found to display hypertension (Levitt, et al. 1996; Langley-Evans,

1997). Chronically elevated plasma glucocorticoid concentrations are associated

with increased susceptibility to hypertension, as well as irnmunosuppression and

diabetes (Baxter, et al. 1987; Munck, et al. 1984; Walker and Williams, 1992).

Studies undertaken in rats have generated important information. Unfortunately,

this information is sornewhat diff~cult to correlate to the situation in the human,

due to the postnatal profile of neuroendocrine maturation in the rat (Matthews.

2000).

Neuroendocrine maturation in the guinea-pig occurs during late fetal life

(Matthews, 1998), which is similar to the pattern of neuroendocrine development

Page 72: Petar - University of Toronto

in primates (Dobbing and Sands, 1973; Dobbing and Sands, 1979). Studies

undertaken in guinea-pigs demonstrated that fetal exposure to synthetic

glucocorticoids during critical periods of brain development. modifies the

developing GR and MR systems in the hippocampus. The alteration of GR and

MR expression within the hippocampus was highly sex-specific. In addition.

modifications at the hippocampal level were accompanied by altered basal

plasma cortisol levels. These alterations were also highly sex-specific (Dean and

Matthews. 1999; Matthews, et al. 2000). It is unclear whether glucocorticoids act

directly at the receptor or at other sites in the brain to influence GR and MR

levels.

Serotonin is known to be involved in maturation and regulation of the HPA

axis (Dinan, 1996). It is well established that serotonin can regulate GR

expression and binding, both in vivo and in vitro (Mitchell. et al. 1990; Mitchel, et

al. 1990). Administration of serotonin to rat hippocampal neurons, in vitro, causes

long-terni increaseç in GR expression as well as binding (Mitchel, et al. 1990).

However, it remains unclear whether this change occurs in neurons andlor glia.

Work described in the previous chapter demonstrated that changes in GR mRNA

were neuron specific.

In Chapter 2 we described glucocorticoid and serotonin-induced changes

in the glucocorticoid receptor mRNA in the mouse. In this chapter we

investigated similar questions but in the more advanced model.

Page 73: Petar - University of Toronto

4.2 Objectives

The objective of this study was to investigate the direct effects of

endogenous (cortisol) and synthetic glucocorticoid (dexamethasone) as well as

serotonin on GR and MR mRNA levels within fetal guinea-pig hippocampal

neurons, in vitro. Based on evidence from in vivo studies, it was decided to

investigate males and females separately. We used a technique that allowed us

to examine GR mRNA and MR mRNA expression in a cell-specific manner.

4.3 Hypothesis

We hypothesized that exposure to cortisol and dexamethasone will down

regulate GR mRNA expression in the fetal guinea-pig hippocampal neurons. We

expected that exposure to serotonin would up regulate GR mRNA expression in

hippocampal neurons. We hypothesized that exposure to cortisol will down

regulate MR mRNA expression, while dexamethasone and serotonin treatments

will not affect expression of MR mRNA within fetal hippocampal neurons.

Although corticosterone did not affect MR mRNA expression in the previous

study (Chapter 3), here we anticipated an effect due to species-specific

differences. Our hypothesis is based on the same rationale as described in

Chapter 3.

Page 74: Petar - University of Toronto

4.4 Methods

4.4.1 Development of Fetal Guinea-pig Culture System

At the onset of my studies, methods for producing cultures of hippocampal

neurons from fetal guinea-pigs were not developed. Therefore. the first step

towards establishing a working protocol involved modification of existing culture

methods, from mouse studies. Applying the mouse culture protocol to fetal

guinea-pig culturing proved to be unsatisfactory, as this did not produce viable

neurons. Further modification of the existing mouse culture protocol involved

modification of culture media, such that culture media contained human brain

extract N2 (Gibco, see Appendixl) and/or neme growth factor (mNGF,

Appendixl). However, combinations again failed to produce viable neurons.

Magistretti et al (1 996), successfully developed adult guinea-pig cortical neuron

culture using supplemental basic fibroblast growth factor (bFGF) . Addition of

bFGF to Our system was not possible due to the prohibitive high cost of bFGF

and the high volume of media required for our experiments.

Examination of fetal guinea-pig hippocarnpal cultures revealed that

supporting non-neural cells were absent from the preparations. Glial cells are

thought to be a source of neurotropic substances and, in practice, most protocols

for culturing CNS neurons depend on high ceIl densities and the rapid

proliferation of endogenous glia to support neuronal survival and development

(Banker and Goslin. 1991). In order to resolve this problem, we modified an

existing Ban ker culture technique (Cultu ring Nerve Cells, 1 991 , pz61 ) which

wmbined mouse cerebellar cells acting to condition the medium for the guinea-

Page 75: Petar - University of Toronto

pig hippocampal cells. The two-type culture system was achieved by seeding

cerebellar cells on the bottom of a petri dish (35mm), and placing guinea-pig cells

on glass cover-slips (25mm diameter, Appendixl ). Cover-slips were mounted

with small paraffin legs, which allowed the cover-slips to be elevated and out of

contact with cerebellar cells. This system produced the first viable fetal guinea-

pig hippocampal neurons in culture. The first group of hippocampal cultures,

derived from gestational day 40 fetuses was undertaken using this system.

4.4.2 Mouse Cerebellar Culture

Accurately time-dated CD1 mouse neonates were obtained from Charles

River Laboratories (St-Constant, Quebec, Canada). Six day old neonates were

euthanized by decapitation. Neonatal heads were disinfected in 70% ethanol. for

5 minutes. Neonatal brains were removed using standard dissection methods

(described in Chapter 2) and placed in dissection medium (see Appendix 1).

Neonatal cerebella were dissected from neonatal brains. At this stage of mouçe

development. cerebella were large enough so that dissection could be perfoned

without the dissection microscope. The cerebella were cut into small pieces.

Dissection medium was removed and replaced with trypsinlEDTA ( l x ) solution

(see Appendix 1) and tissue pieces were incubated for 15 minutes at 37°C.

TrypsinlEDTA solution was removed and tissue was placed in MEM (1.5ml)

containing fetal bovine serum (FBS) and home serum (HS) (see Appendix 1).

Tissue was further dissociated by trituration as described in Chapter 3.

Dissociated tissue was suspended in MEM containing FBS and HS (25ml).

Page 76: Petar - University of Toronto

Cell suspension (0.5ml) was then placed on each collagen-coated 35mm

petri dish containing MEM with FBS and HS (Iml). 50 petri disheç were

generated in total. Each cerebellar culture was allowed to develop for at least five

days prior to initiating guinea-pig culture. 24 hours pnor to initiating guinea-pig

culture, medium was removed from each cerebellar culture and replaced with

MEM containing N2 and mNGF (2.0ml, see Appendix 1).

4.4.3 Guinea Pig Hippocampal Culture, Gestational Day 40

Accurately time-dated pregnant guinea pigs were generated according to

established methods (Elvidge, 1972). Animals were sacrificed on day 40 of

gestation, by decapitation. Fetuses were rapidly removed and decapitated. Fetal

brains were isolated and placed on ice. Gender of fetuses was established by

visual identification of testes or ovaries, upon dissection of the lower abdomen.

Fetal brains from each litter were grouped according to sex and hippocampi

isolated from each brain. Each brain was positioned dorsally and the cerebellum

and frontal cortex were removed. A cut was made on each side of the brain,

between rnidbrain and the two hemispheres. Care was taken not to cut too

deeply, as this would damage the hippocampus. The midbrain was gently

pushed forward and hemispheres were flattened, allowing visualization of the

hippocampus. Each side of the hippocampus was dissected with careful attention

to rernove any non-hippocampal brain tissue. Each fetal hippocampus was

placed in dissection medium (see Appendix 1) and was cut into smaller pieces.

Dissection medium was replaced with trypsinIEDTA (lx). Tissue was incubated

Page 77: Petar - University of Toronto

at 37'C for 20 minutes. Trypsin EDTA was removed and tissue pieces placed in

15ml falwn tubes mntaining MEM-N2 (1.5ml, see appendixl). Tissue was

rnechanically processed with trituration, first passing it through a Pasteur pipette

(10 times) and then through flame-polished Pasteur pipette (10 tirnes).

Dissociated tissue was combined with MEM-N2 (1.5m1, final suspension volume

3ml). Cell suspension (120 PL) was placed on each poly-D-lysine-pretreated

cover slip and allowed to stand in the incubator (37'C) for 40 minutes. Cover

slips were transferred into petri dishes (35mrn) containing mouse cerebellar

cultures. The day of dissection was counted as day 0.

Twenty four hours after the dissection FUDR (200pL, see Appendix 1 ) was

added to each petri dish (as described in Chapter 3). Cultures were then allowed

to incubate for 4 days. On day four the medium was changed. On day six the

cover slips and medium from the culture dishes were transferred into separate

petri dishes (35mm), such that guinea-pig's hippocampal neurons remained in

cerebellar conditioned media but not in the same dishes as cerebellar cells.

Treatment was initiated on day 7. Treatment groups included

dexamethasone, corticosterone and serotonin. Each treatment group was

administered at InM, 10nM and 100nM concentrations, respectively (see

appendix2). Treatments were repeated for 4 days. 24 hours apart. 24 hours after

the last treatment, cultures were fixed with 4% parafomaldehyde (see

appendix3). Cultures were kept under ethanol (95%), at 4"C, until processing

with in situ hybridization.

4.4.4 IN SITU HYBRIDIZATION

Page 78: Petar - University of Toronto

All solutions used in this section were described in detail in Appendix3.

4.4.5 Oligonucleotide Probes

The GR oligonucleotide probe used in guinea pig cultures was

complimentary to bases 1-45 of guinea pig GR mRNA (Keightley et al, 1994).

The MR oligonucleotide probe was complimentary to bases 2942-2986 of human

MR mRNA (Ariza, et al. 1987). Probes were labeled with s~~ as described in

Chapter 3. 80th of the probes have been characterized previously (Matthews.

1998).

4.4.6 Hybridization

Radio-labeled probe was added to hybridization buffer (Appendix 3) so

that final activity in the buffer was 1000 cpmi;iL. Sterile parafilm slips (5cm X

15cm) were placed in incubation chamber. Hybridization buffer (100pL) was

dotted on parafilm. Cover-slips were placed on parafilm face (cells) down. Care

was taken to expel air bubbles undemeath the cover-slips. Moisture levels in the

incubation chamber were maintained with soak solution (Appendix 3). Cover-

slips were incubated overnight at 42OC.

4.4.7 Washing, Emulsions, Developing and Counter-staining Emulsions

Identical to those described in Chapter 3 (see Methods), with the following

exceptions:

Page 79: Petar - University of Toronto

1. Radiosensitive photographic emulsion used was l lford Scientific K.5D. This is

a pre-diluted form of the same emulsion.

2. Counter staining: PBS (2min), methylene blue (0.01%, made in PBS, Imin),

ultrapure water (20s), ethanol (50%, 1 min), ethanol (70%, 2min), ethanol

(95%, 1 min), xylene (3rnin).

4.5 Data analysis

Analysis of GR mRNA and MR mRNA in pyramidal neurons was carried

out at 200X magnification. On average 20-50 grains were identified over positive

neurons and mean grain count per neuron was calculated for each treatment (at

least 4 separate experiments per treatment). Approximately 15 mRNA positive

pyramidal neurons were randomly sampled and analyzed in each well (1

well/treatrnentlanalysis). Cell counting revealed approximately 50 neurons/well.

Non-neural cells were present at very low density or were absent due to the

nature of our culture technique. As a result it was not possible to investigate

mRNA levels in non-neuronal cells.

Group data for each sex are presented as MeanskS.E.M. Additionally,

data for both sexes were pooled and examined together. Data were statistically

analyzed using Statistica (Release 5, 97 Edition, Oklahoma, USA). The effects of

treatment and sex on GR and MR mRNA were deterrnined using a two-way

ANOVA followed by the Duncan's method of post-hoc comparison.

Page 80: Petar - University of Toronto

In the case of lower treatment concentrations (1 and 10nM, GR mRNA

analysis) our n value was too small for analysis to be statistically viable. In those

cases we report trends between data.

MR mRNA analysis was possible only on grouped data from cortisol

treated cells due to a low n value. Analysis of MR mRNA in response to

dexamethasone and serotonin, was not possible even after grouping data. due to

limited material. The reduced MR analysis resulted from extreme difficulty in

handling ernulsion covered wver slips in the dark-room. This led to a heavy loss

of cover slips in Our first (MR mRNA) experiment. Given the time frame of these

studies (1 5 months), it was not possible to repeat these treatments.

4.6 ResuIts

High levels of glucocorticoid receptor mRNA were detected in fetal

hippocampal neurons. Mineralocorticoid receptor mRNA levels were also high in

the fetal hippocampal neurons. Treatment of hippocarnpal cultures with cortisol,

at 1 OOnM, significantly (pc0.05) down-regulated GR mRNA expression in both

sexes (Fig. 4.1a). Exposure to 1 or lOnM did not alter GR mRNA when male and

female data was combined. There was no difference in MR mRNA between

cortisol treatrnent (100nM) and controls (fig.4.l b).

Dexamethasone exposure (100nM) significantly (pc0.05) down-regulated

GR mRNA levels within hippocampal neurons, in both males and females (Fig.

4.2). GR mRNA levels were not different between dexamethasone (1 and 10nM)

Page 81: Petar - University of Toronto

treatment groups and control. Analysis of MR mRNA was not possible due to

problems outlined above.

Serotonin exposure (1 00n M) resulted in significant (~~0.05) up-regulation

of GR mRNA levels within neurons (Fig.4.3). Analysis of MR rnRNA was not

possible due to low n value.

4.7 Discussion

The present study has revealed, for the first time, that glucocorticoid

exposure changes GR mRNA levels in neurons. Both synthetic glucocorticoid

(dexarnethasone) and endogenous glucocorticoid (cortisol) exposure (1 OOnM)

decreased GR mRNA levels in hippocampal neurons, removed from male and

female fetuses at d40 of gestation. Serotonin exposure (100nM) increased GR

mRNA levels in the hippocampal neurons in both sexes.

Previous studies in the guinea-pig have demonstrated that fetal exposure

to glucocorticoids leads to long-term changes in HPA function (Dean, et al. 2000;

Matthews, et al. 2000). Exposure of fetal guinea-pigs to dexamethasone on days

50 and 51 of gestation results in prepubertal male offspring with increased basal

adrenocortical activity (Dean, et al. 2000). The increase in basal adrenocortical

activity is associated with an increase in hippocampal GR mRNA levels (Dean, et

al. 2000). In addition, our laboratory recently demonstrated that fetal exposure to

dexamethasone on days 40, 41, 50, 51, 60 and 61 of gestation results in adult

female offspring with increased basal adrenocortical activity and adult male

offspring with decreased basal adrenocortical activity. Changes in adult female

Page 82: Petar - University of Toronto

offspring were associated with a decrease in hippocampal MR mRNA and a

decrease in PVN and pituitary GR mRNA (Matthews, et al. 2000). Taken together

with evidence from rat studies (Levitt, et al. 1996) these data suggest that the

"programming" of HPA activity is linked to a change in glucocorticoid feedback

sensitivity.

Results from the present study suggest that there are acute effects of

glucocorticoids on GR production in cultured fetal hippocampal neurons but no

direct effect on MR production. It is difficult to establish whether the acute effects

of glucocorticoids on GR synthetic processes are permanent. due to limited life

span of fetal neurons in culture.

Down-regulation of GR mRNA expression following synthetic

glucocorticoid exposure has been reported in other in vitro culture systems using

immortalized cell lines (Cidlowski. et al. 1981 ; Svec and Rudis, 1981; Webster

and Cidlowski. 1994). Permanent down regulation of GR mRNA levels in HeLa

cells was associated with modification in the GR prornoter (Silva, et al. 1994). It

rernains to be investigated whether the same mechanism is responsible for

down-regulation of GR mRNA in developing neurons.

Dean and Matthews (1 999) demonstrated that fetal dexamethasone

exposure in the guinea-pig. on days 50 and 51 of gestation. results in an

increase of hippocampal GR and MR mRNA levels in the fernale fetus, on day

52. The same study found no change in hippocampal GR and MR mRNA in the

male fetus. The acute effects of dexamethasone in that study differ frorn the

present findings. The reasons for this discrepancy are unknown at present.

Page 83: Petar - University of Toronto

However, it is possible that in in vivo studies dexamethasone also affects the

maturing serotonergic system, which is involved in the hippocampal GR

regulation. Dexamethasone has been found to promote maturation of

serotonergic systems (Slotkin, et al. 1996). ln the guinea-pig, dexamethasone

exposure was found to induce an acute increase in fetal thyroid hormone (Dean

and Matthews, 1999), which is known to increase hippocampal serotonin

turnover in the rat (Meaney, et al. 1987). If this hypothesis is correct then it

appears that direct effects of gluwwrticoids on the hippocampal neuron GR

mRNA levels are masked or ovemdden by the effects of increased serotonergic

activity. However, this remains to be investigated further. One possible way of

examining this issue would be to treat neurons with dexamethasone/serotonin

combination and analyze subsequent changes in GR mRNA. However, it is also

possible that glucocorticoids may affect other additional neurotransmitter

systems that modify GR compliment. It has been demonstrated that alterations in

hippocampal norepinephrine can mode@ corticosteroid receptor levels

(Barbazanges, et al. 1996). Slotkin and wlleagues demonstrated that

norepinephrine turnover in the cerebellum and forebrain is reduced following

dexamethasone exposure in utero (Slotkin, et al. 1992). Norepinephrine content

within the hippocampus and neocortex was also reduced following

dexamethasone exposure in ufero (Muneoka, et al. 1 997). Longitudinal studies

have shown that in utero glucocortiwid exposure results in premature maturation

of norepinephrine systems in the brain stem, forebrain and cerebellurn (Slotkin,

et al. 1992) and results in overexpression of norepinephrine transporter (Slotkin,

Page 84: Petar - University of Toronto

et al. 1991). It is clear that dexamethasone profoundly influences the

development of norepinephrine system. Unfortunately, the long-terni

consequences (on HPA function) of the norepinephrine system manipulation are

not known. Future studies examining long term-effects of norepinephrine system

on the HPA function are required. To conclude, it is possible that differences

observed in in vivo situation result from the norepinephrine system input.

Previous studies reported sex-differences in hippocampal GR and MR

mRNA levels, as a result of dexarnethasone treatrnent (Dean and Matthews,

1999; Dean, et al. 2000; Matthews, et al. 2000). It was suggested that male and

female HPA axis and corticosteroid receptor systems are at different stages of

development at the time of exposure, and thus, respond differently (Dean, et al.

2000). In the present study, sex-differences in the hippocampal GR mRNA, in

response to direct dexamethasone exposure, were not observed. GR mRNA

levels were found to decrease in response to dexamethasone exposure. in both

male and female derived hippocampal neurons. There are several possibilities

that may explain this observation. In the male fetal neurons, circulating

testosterone is converted to estrogen. This is mediated by the enzyme P450

aromatase (MacLusky, et al. 1987). Activity of P450 is believed to be responsible

for sexual differentiation observed in the hippocarnpus and CNS in general

(MacLusky, et al. 1987). The effects of estrogen in the developing CNS have

been suggested to be mediated by estrogen receptors (O'Keefe and Handa.

1990). Therefore, in the male fetal neurons, sex hormones and dexamethasone

could act in synchrony to modrfy GWMR levels within the hippocampal neuron.

Page 85: Petar - University of Toronto

Both sex hormones and glucocorticoids act through similar transcriptional

machinery (Barnberger, et al. 1996) and it is possible that they interact. It would

be interesting to conduct a study where hippocarnpal neurons in culture are

treated with combinations of both testosterone/dexamethasone or

estrogenldexamethasone. This would establish whether the observed differences

between in vivo and in vitro studies in the male fetuses, are due to the absence

of sex hormones in the latter system. The second possibility involves sex

hormone modification of certain systems, which like the serotonergic system,

influence the hippocampal GWMR populations. Presence of sex hormones could

increase or suppress the maturation of the neurotransmitter system in question.

Alteration in maturation could change the activity of this system, and therefore

influence development of hippocampal GR and MR populations. This remains to

be investigated. Finally. the observed differences between the two systems could

be explained by the differences in dexamethasone concentration. It is possible

that 100nM dexarnethasone applied in in vitro system is higher than

concentration of dexamethasone experienced at the hippocampus in in vivo

studies. Also, distribution of dexamethasone in in vivo studies could be affected

by the presence of P-glycoproteins within blood-brain bamer, which have the

potential to exclude dexamethasone from the brain (Meijer, et al. 1998).

The present study did not detect glucowrticoid-induced modifications in

hippocampal MR mRNA levels. This finding implies that the long-term effects of

glucowrticoid exposure demonstrated in female adult offspring (Matthews, et al.

2000) occurç through an indirect mechanism. Corticosterone has 10-fold higher

Page 86: Petar - University of Toronto

affinity towards MR than GR (Rupprecht, et al. 1993; Anna, et al. 1988; Evans

and Anna. 1 989; Cato, et al. 1 991 ).

Prenatal programming of the HPA axis occurs, at least partially, at the

level of hippocarnpal GR and MR (Matthews. 2000). Hippocampal GR and MR

are involved in regulation of HPA activity (De Kloet. et al. 1998). Evidence

indicates that the MR is involved in feedback regulation of basal HPA function

while the GR is important during periods of HPA activation (De Kloet, et al.

1998). If similar GR autoregulation, to that which we report in vitro, occurs in vivo,

then this may itself have very significant consequences for normal hippocampal

development.

The role of serotonin in the regulation of GR development in primary

hippocampal culture, as weil as importance of thyroid-serotonin system in

regulating hippocampal GR levels during development. was addressed in

Chapter 2. In the present study. four days of serotonin exposure (100nM)

increased GR mRNA levels in the hippocampal neurons. It remains to be

detemined whether this increase is permanent. This finding could exptain the

acute increase in GR mRNA, which is documented in the female guinea-pig

fetuses. on 52"d day of gestation (Dean and Matthews, 1999). The same study

reported acute increase in fetal thyroid hormone in response to dexamethasone

administration. It is likely that the serotonergic system is mature by the time of

exposure, and is thus, able to respond to increased thyroid hormone secretion by

increasing serotonin tumover. lncreased serotonin tumover could, in tum,

increase hippocampal GR mRNA levels. This effect was rewrded in the rats

Page 87: Petar - University of Toronto

(Meaney, et al. 1987). and discussed in Chapter 2. The findings of this study

support a view that serotonin plays an important role in the prograrnming of

hippocampal corticosteroid receptors, and indicate a direct effect of serotonin on

hippocampal neurons.

To conclude, results of this study indicate that exposure to glucocorticoids

has a direct effect on GR mRNA expression in fetal guinea-pig (gestational day

40) hippocampal neurons. In addition, we have shown that exposure to serotonin

increases the GR mRNA complement within hippocampal neurons.

Findings described in this chapter were recorded in neurons which were

denved from fetuses at 40 days of gestation. In the guinea-pig, neurogenesis is

almost completed by gestational day 40 (Dobbing and Sands. 1979). Studies

undertaken in vivo reported changes in hippocampal GR and MR mRNA in

fetuses at 50 days of gestation (Dean and Matthews. 1999). It is established that

the most rapid period of brain growth in the guinea-pig occurs by gestational day

50. The rapid phase of brain growth is charactenzed by rapid neuronal

proliferation, synaptogenesis, dendritic aborization and increase in brain weight

(Dobbing and Sands. 1979). The period of rapid brain growth is also associated

with rapid neuroendocrine development (Dobbing and Sands.1979). Differences

in responses to dexamethasone between in vivo and in vitro studies could be

attributed to different stages of neuronal development. In the hippocampus, GR

mRNA is expressed at higher levels at gestational day 50 than at gestational day

40 (Matthews. 1998). It is possible that neurons, at 50 days of gestation, are

Page 88: Petar - University of Toronto

more responsive to dexamethasone due to higher GR levels. This possibility was

investigated in Chapter 5.

Page 89: Petar - University of Toronto

Figure 4.1 :

A) analysis of GR mRNA following cortisol (O. 1. 10, 1OOnM) exposure (4 days, n=6). Results are

expressed as mean+S.E.M. Yndicates statisücal difference from control (pcO.01).

(Numbers under bars represent the n value for each group; c=combined data; f= females only.

n=6; m= males only, n=6)

5) Analysis of MR mRNA following cortisol (O, 1 OOnM) exposure (4days, n=6). Results are

expressed as meankS.E.M. Analysis was perfomed on grouped data (males + females) only.

Page 90: Petar - University of Toronto

Figure 4.1

c m f c m f c m f c m f

O 1 10 100

Treatrnent (nM)

O 1 O0 Treatment (n M)

Page 91: Petar - University of Toronto

Figure 4.2:

Analysis of GR mRNA levels following dexamethasone (O, 1, 10, 100nM) exposure (4 days).

Results are expressed as meankS.E.M. 'Indicates statistical defierence from control (p<0.01).

(Numbers under bars represent the n value for each group; C=cornbined data; m=males alone.

n=6; f=fernales alone, n=6)

Figure 4.3:

Analysis of GR mRNA levels following serotonin (O, 1, 10, 100nM) exposure (4 days). Results are

expressed as mean2S.E.M. *Indicates statistical difference from wntrol (p<0.01).

(Numbers under bars represent the n value for each group; C=combined data; m=males alone,

n=6; Hernales alone, n=6)

Page 92: Petar - University of Toronto

Figure 4.2

1 .*. -.

... *.- W . . . -.

S . . ... ... ... *:.:.: . *. S . . .*. ...

t r . ... ... ... ... ... 2

c m f c r n f c m f c m f

O 1 I O 100

Treatment (nM)

Figure 4.3

c r n f c m f c m f c rn f

1 I O IO0 Treatment (nM)

Page 93: Petar - University of Toronto

CHAPTER 5

Regulation of

Development:

Days

5.1 Introduction

Hippocampal GR and MR mRNA During

Guinea-pig Studies, Gestational Age 50

Studies in Chapter 4 described corticosteroid mRNA alterations in fetuses

at 40 days of gestation. In the guinea-pig, days 40 and 50 of gestation are

considered cntical times for brain development (Dobbing and Sands. 1979).

Exposure to glucocorticoids in the human would likely occur during similar critical

times in human brain development. Guinea-pigs give birth to neuroanatomically

and neuroendorinologically mature Young. Peak brain growth. in the guinea pig,

occurs between 50 and 52 days of gestation (-75% of gestation; gestation length

- 70 days) (Dobbing and Sands, 1970; Dobbing and Sands. 1979). The rapid

phase of brain growth is characterized by rapid neuronal proliferation,

synaptogenesis, dendritic aborization and increase in brain weight (Dobbing and

Sands. 1979). The period of rapid brain growth is also associated with rapid

neuroendocrine development (Dobbing and Sands.1979). Neuroendocrine

maturation and development of central GR and MR systems occurs during Iate

fetal life in this species (Matthews, 1998). GR and MR populations go through

rapid and neuron-specific changes during this period. These changes can be

acutely and pemianently modified by fetal glucowrticoid exposure (Dean and

Page 94: Petar - University of Toronto

Matthews. 1999; Dean, et al. 2000; Matthews, et al. 2000). Cleariy, gestational

day 50 represents a critical period in the development of a guinea pig brain. This

period is wnsidered to be particularly vulnerable to drugs and other

pathophysiological insults (Dobbing and Sands. 'l979).

5.2 Objectives

Acute changes in hippocampal GR and MR mRNA levels at gestational

day 52, as a result of fetal dexamethasone exposure on days 50 and 51 of

gestation, were demonstrated previously, in vivo (Dean and Matthews, 1999). It

remained unclear whether these changes resulted from direct actions of

glucocoriicoids on the hippocampal neurons. The main objective of this study

was to investigate the direct effects dexarnethasone on hippocampal GR and MR

mRNA levels, in vitro. Direct effects of serotonin on hippocarnpal GR mRNA and

MR mRNA were also examined. Effects of cortisol were not examined in this

study.

5.3 Hypothesis

We hypothesize that exposure to dexamethasone will down regulate GR

mRNA expression in fetal guinea-pig hippocampal neurons. Exposure to

serotonin will up regulate GR mRNA expression in hippocampal neurons. We

hypothesize that exposure to dexamethasone or serotonin will not affezt

expression of MR mRNA within fetal hippocampal neurons. The rationale for this

hypothesis is identical to one described in Chapters 3 and 4.

Page 95: Petar - University of Toronto

5.4 Methods

5.4.1 Development of guinea-pig culture system

During the processing of fetal guinea-pig cultures it was establiçhed that

further optimization of the system was required. Manipulation of cells in this

system, such as media exchange or treatment was time consuming and it

increased the risk of contamination during handling. In addition, the cover-slips

were difficult to handle dunng N, situ hybridization analysis. We also had to

design and manufacture storage containers and cover-slip racks in the lab. The

greatest difficulty was experienced during emulsion application and exposure,

during which many of the slides adhered to each other and hence becarne

unusa ble.

We proceeded to further optimize our culture system, especially focusing

on final in situ handling. We adopted the use of tissue culture inserts (IOmm, Life

Technologies, Toronto, Canada), which allowed us to use the chamber slide

system similar to that desct-ibed for the mouse studies. This resolved al1 technical

difficulties associated with final in situ processing and analysis. In this system,

mouse cerebellar cells were grown on the insert and guinea-pig cells were grown

on the glass bottom of the chamber slide. This system produced viable fetal

guinea-pig (gestational day 50) neurons and was very easy to use.

5.4.2 Mouse Cerebellar Culture, On Culture lnserts

Preparation of the mouse cerebellar cultures was identical to that

described in Chapter 4, with the following difference:

Page 96: Petar - University of Toronto

1. Cell suspension (0.1 -0.2rnl) was placed in each culture insert.

5.4.3 Guinea Pig Hippocampal Culture, gestational day 50

Preparation of the guinea-pig hip pocam pal cells was identical to that

descnbed in Chapter 4, with the following differences:

1. Cell suspension (250pL) was placed in each PDL-precoated well. Each well

previously contained MEM-N2, mNGF (1 750pL). Culture-inserts containing

mouse cerebellar culture were inserted into the each well. Total of 16

welIs/sexfanimal were prepared.

2. Each culture slide well contained cuIture insert and 1 ml of MEM-FBS, HS.

5.4.4 IN SITU HYBRIDIZATION

All solutions used in this section were described in detail in Appendix3. In

situ hybridization and subsequent ernulsion autoradiography was identical to that

descnbed in Chapter 3. The same anti-sense oligonucleotide probes as those

described in Chapter 4 were used.

5.5 Data analysis

Analysis was sirnilar to that carried out in Chapters 3 and 4. Analysis of

GR mRNA and MR mRNA in pyramidal neurons was carried out at 200X

magnification. On average 20-50 grains were identiied over positive neurons and

mean grain count per neuron was calculated for each treatrnent (4 separate

experiments per treatment). Approximately 30 mRNA positive pyramidal neurons

Page 97: Petar - University of Toronto

were randomly sampled and analyzed in each well (1 well/treatment/analysis).

Cell wunting revealed approximately 70 neurons/well. Non-neural cells were

present at very low density or were absent due to the nature of Our culture setu p.

Given the lack of sex difference observed in Chapter 4, statistical analysis

of pooled (male + fernale) cultures was undertaken though male and female data

are also presented. Group data for each sex are presented as MeanskS.E.M.

Data were statistically analyzed using Statistica (Release 5, 97 Edition,

Oklahoma, USA). The effects of treatment on GR and MR mRNA were

detemined using a two-way ANOVA followed by the Duncan's method of post-

hoc corn parison.

5.6 Results

High levels of glucocorticoid receptor mRNA were detected in

hippocampal neurons removed at 50 days of gestation. Mineralocorticoid

receptor mRNA levels were also high in the hippocarnpal neurons.

Dexarnethasone treatment (1 00nM) significantly ( ~ ~ 0 . 0 5 ) down-regulated

GR mRNA levels within neurons (Fig. 5.la). Dexamethasone treatment at lower

concentrations (1 and 10nM) did not change GR mRNA levels in neurons

(Fig.5.1 a). MR mRNA levels were not affected by dexarnethasone treatment, at

any of the tested concentrations (1, 10 and 1 OOnM)(Fig.5.1 b).

Treatment of hippocampal cultures with serotonin, at 1 OOnM, significantly

( ~ ~ 0 . 0 5 ) upregulated GR mRNA expression (Fig.5.2a). Exposure to serotonin. at

1 and IOnM, did not alter GR mRNA levels (Fig.5.2a). Serotonin administration

Page 98: Petar - University of Toronto

had no impact on MR mRNA levels, at any of the concentrations tested

5.7 Discussion

Results from this study are consistent with those reported in neurons

derived from 40 days old fetuses (Chapter 3). Dexamethasone exposure

decreased GR mRNA levels in hippocampal neurons, while serotonin exposure

increased GR mRNA levels. In the present study we describe acute effects on

GR mRNA. It remains to be established whether synthetic processes are affected

permanently.

Effects of dexamethasone in this study contrast those reported in in

vivo situation. Dexamethasone exposure, in vivo, induced up-regulation of GR

mRNA within fernale hippocampal neurons and it had no effect on GR mRNA

expression within male hippocampal neurons (Dean and Matthews. 1999).

Discrepancies between the two studies can be explained by two possibilities. It is

possible that presence of serotonergic system in vivo, as well as the presence of

noradrenergic systems that also modifies GR compliment, counter-acted the

negative input of glucocorticoids. As it was discussed in the previous chapter,

glucocorticoids enhance the maturation of serotonergic system, whose activity is

known to cause up-regulation of GR mRNA within the hippocampus. Based on

the findings in neurons derived from animals at 40 and 50 days of gestation, one

can suggest that actions of serotonergic system, h vitro, are more potent than

the direct actions of glucocorticoids. It would be interesting to test this proposal in

Page 99: Petar - University of Toronto

vitro, in neurons derived from animals at 50 days of gestation. Information

generated at this gestational age, in combination with similar information from 40

days of gestation. would greatly enhance Our understanding of serotonergic

system input in HPA programming during development. The second possibility

for observed discrepancies in two studies is glucocorticoid concentration

difference in the hippocampus. It is possible that glucocorticoid concentration in

the hippocampus, in vivo, is too low to effect GR mRNA expression. In in vivo

studies distribution of dexamethasone could be affected by the presence of P-

glycoproteins within blood-brain bamer. which have the potential to exclude

dexamethasone from the brain (Meijer. et al. 1998).

Dexamethasone has a very low affinity for MR and it selectively binds GR

(Veldhius, et al. 1982). Therefore. it is not surprising that the dexamethasone

treatment failed to affect MR mRNA levels. Serotonin exposure did not modify

MR mRNA levels in neurons. This was similar to the situation in neurons derived

from gd40 fetuses. The absence of serotonin effect on MR protein levels, in vitro,

was recorded in rat studies (Mitchell, et al. 1990). Taken together, these latter

findings suggest that development of rnineralocorticoid receptor system is not

influenced by serotonin at 50 days of gestation in the guinea-pig. Serotonin

administration u p-regulated GR mRNA expression. This finding is in agreement

with results previously reported in mouse studies and gestational day 40 guinea-

pig neurons. Taken together with studies done in rats (Meaney, et al. 1987;

Meaney et al. 1994), these findings support the involvement of thyroid-serotonin

system involvement in HPA programming, as discussed in Chapter 2.

Page 100: Petar - University of Toronto

Results described in gestational day 50 neurons were identical to those in

neurons that were derived from animals at 40 days of gestation. There was no

difference in response to dexamethasone or serotonin despite the major

differences in hippocampal development at these times. Based on these findings

one can propose that the main direct effect of glucocorticoids, on fetal neurons, is

to down-regulate expression of GR. It would be interesting to test this further, in

neurons derived from animals that are closer to term. However, such neurons

would be difficult to culture due to advanced level of neuroanatomical maturity in

the guinea-pig fetuses which are close to ten .

In surnmary, results of this study indicate that exposure to glucocorticoids

has a direct effect on GR mRNA expression in fetal guinea-pig hippocampal

neurons. Furthenore, this study has shown that exposure to serotonin increases

the GR mRNA complement within hippocampal neurons. One can speculate that

both glucocorticoids and serotonin exert their influence on the developing

hippocampus, but that the net effect of such treatment is dependent on the

maturational stage of both systems, at the time of exposure. However, the main

direct effect of glucocorticoids, on fetal neurons, is to down-regulate expression

of GR.

Page 101: Petar - University of Toronto

Figure 5.1:

A) Analysis of GR mRNA following dexamethasone (O, 1, 10, 100nM) exposure (4 days). Results

are expressed as rnean~S .E.M . 'Indicates statistical difference from control(p<O.Ol).

(Numbers under bars represent the n value for each group; C=combined males+females; m=males

alone; n=3; Hemales alone; n=2)

B) Analysis of MR mRNA following dexarnethasone (O, 1, 10, 100nM) exposure (4 days). Results

are expressed as rneankS.E.M.

(Numbers under bars represent the n value for each group; C=combined males+females; m=males

alone; n=3; Memales alone; n=2)

Page 102: Petar - University of Toronto
Page 103: Petar - University of Toronto

Figure 5.2:

A) Analysis of GR mRNA following serotonin (O, 1, 10, 1 OOnM) exposure (4 days). Results are

expressed as mean6.E.M. 'Indicates statistical difference frorn control(p<O.Ol).

(Numbers under bars represent the n value for each group; C=combined males and females;

m=males alone; n=3; f=fernales alone; n=2)

B) Analysis of MR mRNA following dexamethasone (O, 1, 10, 100nM) exposure (4 days). Results

are expressed as meantS.E.M.

(Nurnbers under bars represent the n value for each group; C=combined males and females;

m=males alone; n=3; Hernales alone; n=2)

Page 104: Petar - University of Toronto

Figure 5.2

A 40 *

s mg L 30 m u

a 2 20 ar E P! I O C3

O I 1

c ; ? 9 1 5 ' 31 2l

c m f c m f c m f

1 I O 1 O0

Treatment (nM)

L

c r n f

O

c r n f c m f c m f c m f

O 1 10 100 Treatment (nM)

94

Page 105: Petar - University of Toronto

CHAPTER 6

Multi-drug Resistance Studies

6.1 Introduction

The hippocampus is one of the most sensitive targets for the action of

endogenous and synthetic glucocorticoids. Excess glucocorticoid is detrimental

to neuron development (Uno, et al. 1994; Uno, et al. 1990; Matthews. 2000). In

adults, it has been çuggested that dexamethasone (a synthetic glucocorticoid)

penetrates poorly into the brain due to expression of the multidrug resistance

gene (MDRl a) (Meijer, et al. 1998). Drug-transporting P-glycoprotein, a product

of the multi-drug resistance gene, functions as an exclusion pump for synthetic

glucocorticoids and other chemotherapeutic agents (De Kloet, et al. 1998;

Schinkel, et al. 1995; Meijer, et al. 1998). Approximately 10% of pregnant women

in North America, at risk of pre-term delivery, are treated with synthetic

glucocorticoids in late gestation to promote fetal lung maturation (NIH, 1995).

However, little is known about the effects of dexamethasone on fetal brain and

there is no information concerning the expression of MDRI a in the developing

brain. It is likely that exposure of the fetal brain to synthetic glucocorticoids could

be regulated by expression of the MDRla gene.

Page 106: Petar - University of Toronto

6.2 Objectives

The primary objective of this study was to examine the expression of

MDRl a gene in the guinea-pig brain throughout development. The secondary

objective of this study was to detemine whether two-day dexamethasone

exposure in fetal life alters the expression of MDRl a gene.

6.3 Hypothesis

The MDRla gene is expressed in the fetal brain but expression changes

during development. Glucocorticoid exposure duting fetal life can alter the

expression of MDRl a gene.

The first part of Our hypothesis was based on the fact that MDR

expression in adult anirnals was found to protect against dexamethasone

exposure (Meijer et al. 1997). It is possible that the MDR expression in fetuses

could have the same effect. If the MDR expression is to protect the brain during

critical petiods of development, the expression of this gene should have a simiiar

pattern to that of a guinea-pig brain development. The second part of our

hypothesis was based on findings of in vitro studies which demonstrated that

certain P-gp su bstrates, like vinblastine or rnetkephamid, induced or su ppressed

MDR expression (Meijer, et al. 1997).

Page 107: Petar - University of Toronto

6.4 Methods

6.4.1 Animai Work

All animal tissues used in this section were generated previously by S.G.

Matthews and F. Dean. Accurately tirne-dated guinea pigs (Elvidge, 1972; Ree

and Hounslow, 1971) were killed on days 40-45, 50-55 and 60-65 of gestation.

Fetal brains were rapidly removed and frozen. A group of 7 day old neonates and

adult animals were also killed and brains were collected. Each group comprised

six to seven animals.

An additional group of pregnant guinea pigs was subcutaneously injected

with dexamethasone (Imglkg of body weight; n=9) or vehicle (200pL;

n=lO)(Dean and Matthews, 1999). Injections were given at 0800h on days 50

and 51 of gestation. Pregnant guinea pigs were killed on day 52 and the fetal

brains were rapidly removed and frozen.

6.4.2 Tissue Preparation and Sectioning

Frozen guinea-pig brains were coronally sectioned (12pm) using a

cryostat (JUNG CM3000, Leica instruments GmbH, Nussloch, Germany).

Sections were thaw-mounted on poly-L-lysine coated glass rnicroscopic slides.

Sections were stored at -80°C. Prior to hybridization the sections were fked in

4% paraformaldehyde solution (5min). Slides were washed in phosphate buffer

saline (PBS) (2Xlmin) and dehydrated in 70% ethanol (5min) and 95% ethanol

('lmin). This animal work was camed out by F. Dean and S.G. Matthews.

Page 108: Petar - University of Toronto

6.4.3 IN SITU HYBRIDIZATION

The author of this thesis did al1 in situ hybridization work and subsequent

analysis. ln situ hybndization was identical to that described in chapter 3, with the

following exceptions:

1. MDRla oligonucleotide probe hybridized tu guinea pig brain cryo-sections

was complementary to bases 202-246 of Chineçe hamster MDR mRNA. This

sequence is homologous across many rodent species, including that of the

guinea-pig (Croop et al, 1989).

6.5 Autoradiography and Analysis

Slides were dried and exposed to autoradiographic film (Biomax, Eastman

Kodak Co.. Rochester, N.Y., USA) in a light proof cassette. standards

(American Radiochernical Inc., St. Louis, M.O., USA) were exposed along with

the slides to ensure analysis within the linear range of the autoradiographic film.

Cassettes were stored at room temperature. Films were exposed for 28 days and

developed using an automatic processor (Kodak M35A- OMAT processor).

Analysis of autoradiographic signals on film was camed out using a

computerized image analysis system (Imaging Research Inc., St. Catherines,

Ontario, Canada).

MDR mRNA levels were measured in the hippocampus (CA1, CA2, CA3

and CA4 subfields), dentate gyrus, thalarnic nuclei and cortex. Autoradiographic

signal was measured in each area (8 sections per area).

Page 109: Petar - University of Toronto

Statistical analysis was perfotmed using Statistica (Release 5, 97 Edition,

Oklahoma, USA). Data are presented as MeansIS.E.M. and were analyzed

using two-way ANOVA followed by the Duncan's method of post-hoc

comparison. Statistical significance was set at pc0.05.

6.6 Results

MDR1 a mRNA was detected in several regions of the fetal guinea pig

brain. High levels of expression were detected in the CAIICA2 regions of the

hippocampus, CA3 region of the hippocampus, dentate gyrus and thalamic nuclei

(Fig . 6.1 ). Silver emulsion autoradiogra phy performed on coronal sections

revealed cell-specific expression of MDRla mRNA (Fig. 6.2).

Within the CAlICA2 region of the hippocampus, expression of MDRl a

mRNA was detected at al1 gestational groups as well as in neonatal and adult

anirnals. The highest expression was observed in fetuses at 50-55 days of

gestation. Expression of MDRla mRNA progressively decreased towards term. A

significant decrease (pc0.05) in MDRl a mRNA expression was detected

between animals at 60-65 days of gestation and seven day old neonatal animals

(Fig.6.3).

In the CA3 region of the hippocampus, the highest expression of MDRla

mRNA was detected in fetuses at 40-45 and 50-55 days of gestation. Expression

of MDRl a mRNA progressively decreased towards term. There was a significant

reduction ( ~ ~ 0 . 0 5 ) in MDRla mRNA between 50-55 days of gestation and 60-65

Page 110: Petar - University of Toronto

days of gestation. MDRla mRNA levels rernained relatively stable into the

adulthood (Fig.6.4).

In the CA4 region of the hippcampus, the highest expression of MDRla

mRNA was detected in fetuses at 40-45 and 50-55 days of gestation. Expression

of the MDRla mRNA significantly decreased ( ~ ~ 0 . 0 5 ) between 50-55 days of

gestation and 60-65 days of gestation. After 60-65 days of gestation the

expression remained relatively constant at term and through to adulthood (Fig.

6.5).

In contrast, in the dentate gyrus MDRla mRNA levels were detected at

low levels by 40-45 days of gestation. MUR1 a mRNA levels then progressively

increased towards tem. Significant up-regulation ( ~ 4 . 0 5 ) in the MDRla mRNA

levels was detected between 50-55 days of gestation and 60-65 days of

gestation (Fig. 6.6). MDR mRNA levels remained at this increased level in

neonates as well as adult animals.

In the cortex the pattern of MDRla mRNA expression was similar to that

observed in the hippocampus. The highest levels of expression of the MDRla

mRNA were observed at 40-45 days of gestation. Significant (pc0.05) reduction

in the MDRla mRNA expression were detected between 4045 days of gestation

and 50-55 days of gestation as well as between 60-65 days of gestation and 7

day old neonates (Fig. 6.7).

In the anteroventaral thalamic nuciei MDRla mRNA was detected at

gestational days 40-45, 50-55 and 60-65. Expression decreased drarnatically

after t e n and in some animals wuld not be detected (Fig. 6.8).

Page 111: Petar - University of Toronto

Finally, no significant differences were observed in limbic regions,

between animals that were exposed to dexamethasone and animals that

received vehicle alone (Fig. 6.9).

Discussion

Using the MDRla probe, we have described the expression of MDRla

mRNA in the developing brain. We have also shown that levels of MDRla mRNA

change during development in a region-specific manner.

The pattern of MDRla mRNA expression coincides with the pattern of the

guinea pig brain development. The highest levels of MDRla mRNA expression

within the hippocampus and cortex were observed on gestational days 40 and

50. In the guinea-pig, neurogenesis in these areas is completed by day 40 of

gestation, while the most rapid brain growth is observed between days 50 and 55

(Dobbing and Sands, 1970; Dobbing and Sands, 1979). Given the evidence, one

can suggest that MDRla expression is high in order to ensure maximal

protection against xenobiotic substances, during these critical periods of brain

development.

In the dentate gynis, the highest levels of MDRla mRNA expression were

observed juçt prior to term (65 days of gestation) and in neonatal and adult

animals. Development of dentate gynis is known lag behind the hippocampus

(Dobbing and Sands, 1970; Dobbing and Sands, 1979). Hence, it is not

surprising that peak MDRl a expression was observed at a later gestational date.

It is interesting that MDRia mRNA was expressed at very similar levels in

Page 112: Petar - University of Toronto

neonatal and adult animals. This may be explained by the fact that neurogenesis

at low levels, in the dentate gynis, is observed in adult animals (Gould, et al.

1994; Gould, et al. 1991). Continued high expression of MDRl a mRNA within the

dentate gyrus may act to protect neurogenesis from detrimental influences of

xenobiotic agents.

An unexpected finding of this study was the high expression of MDRl a

mRNA within the specific anteroventral thalamic nuclei. In fact, the highest

expression of MDRla mRNA, in gd50 fetal brainç, was recorded in this structure

(see Fig. 10). More perplexing was the dramatic decrease in expression after

t e n and subsequent disappearance of expression in some animals. The high

levels of MDRla mRNA expression in this thalamic nucleus may be partially

explained by its close proximity to the lateral ventricle. High MDRla expression

could protect against the xenobiotic compounds penetrating from cerebrospinal

fluid. However, it is not known how this structure is involved in development.

Such a high level of protection suggests an important role, but this remains to be

investigated .

Since the synthetic glucocorticoids are substrates for the P-glycoprotein

cellular extrusion pump, it is possible that they could induce their own resistance.

The induction of resistance by other P-glycoprotein substrates had been

described in vitro (Anderle, et al. 1998). In this study, two day dexamethasone

exposure (I mglkg of body weight) had no significant effect on the expression of

MDRla gene, in day 50 fetuses. Based on this finding one can speculate that

dexamethasone cannot induce its own resistance. However, this remains to be

Page 113: Petar - University of Toronto

confirmed in studies where dexamethasone would be administered in a repeated

fas hion.

It was previously reported that penetration of dexamethasone into the

brain of MDRla knockout mouse (-1-) is 3-fold higher than that of a wild type

(Schinkel, et al. 1995). The enhanced uptake of dexamethasone in the MDRla (-

/-) brain disappeared when the dose of dexamethasone was raised to lmglkg

mouse, from the previous dose of 0.2mglkg mouse (Schinkel, et al. 1995). This

indicated a limited capacity of the P-glycoprotein transport system. Exposure to

dexamethasone at 100nM (this is approximately 0.014mg) induced down

regulation in GR mRNA within mouse hippocampal neurons (Chapter3). This

dose is higher then that at which the P-glycoprotein saturation is reached

(0.004mg in mouse weighing 209). Therefore, changes in GR mRNA within

mouse neurons due to dexamethasone (1 00nM) exposure could be explained by

P-glycoprotein transport saturation. Absence of effect at lower concentrations of

dexamethasone (1 an 10nM) could be explained by the P-glycoprotein-mediated

exclusion of dexamethasone.

In conclusion, the MDRl a gene is widely expressed in the developing

brain, in a region-specific manner. MDRla gene expression changes during

development in a highly region-specific manner. We speculate that the local

expression of MDRla gene can protect against exogenous glucocorticoid

exposure at low to moderate levels. Expression of the MDRla gene allows

exclusion of dexamethasone from specific regions of the fetal brain and this

could have a protecüve effect during critical periods of development.

Page 114: Petar - University of Toronto

Figure 6.1: Color-enhanced image of MDRla in coronal sections of a fetal guinea pig. following in situ hybridizaüon. MDRla mRNA was present throughout the brain with the folowing regions showing high levels of expression: CAIICA2 region of the hippocampus (CAI), CA3 area of the hippocampus (CA3). dentate gynis (DG) and thalamic nuclei (T).

Legend: dark blue=background; pink=low to moderate expression; red=high expression

Figure 6.2: Images of coronal sections that underwent silver emulsion autoradiography. MDRla mRNA is represented by black clusters of dots.

Page 115: Petar - University of Toronto

rY *- )I Thalamus $

5 al-

*

Hippocampus \ - 2 3 . i-

h - : . 4 <; <:s - - ' - W .

B. Vessel - ~ t ' . - a . - a7

Page 116: Petar - University of Toronto

Figure 6.3: MDRla mRNA levels within CAlICA.2 region of the hippocampus. Highest expression was observed at gd50-55, which corresponds to period of the most rapid brain growth in the guinea pig. Expression significantly (~~0.05) decreased between gestational ages 60-65 and 7 day old neonatal animals.

Figure 6.4: MDRl a mRNA expression within CA3 region of the hippocampus. Expression levels were highest at gd40-45. Expression significantly (~~0.05) decreased between gestational ages 50-55 and 60-65.Expression remained at deceased levels into the adulthood.

Page 117: Petar - University of Toronto

Figure 6.3

40-45 50-55 60-65 7 AduIts

GestationalINeonatal age (days)

Figure 6.4

40-45 50-55 60-65 7 Adults

GestationalINeonatal age (days)

107

Page 118: Petar - University of Toronto

Figure 6.5: MDRla mRNA levels in CA4 region of the hippocampus. Highest expression was observed at gd50, which decreased progressively towards tem. Expression significantly ( ~ 4 . 0 5 ) decreased between gestational ages 50-55 and 60-65.

Figure 6.6: MDRl a mRNA expression within dentate gyms. In contrast to the hipocarnpus, mRNA levels peaked at terni and remained at the same level throughout neonatal life. Expression significantly (pe0.05) increased between gestational ages 50-55 and 60-65.

Page 119: Petar - University of Toronto

Figure 6.5

40-45 50-55 60-65 7 Adults

GestationalINeonatal age (days)

Figure 6.6

40-45 50-55 60-65 7 Adults

GestationallNeonatal age (days)

1 O9

Page 120: Petar - University of Toronto

Figure 6.7: MDRla mRNA expression within the cortex. Expression levels were uniform between the layers. Highest levels were detected at gd40 and then decreased progressively towards term. Expression significantly (p<0.05) decreased between gestational age 4045 and 50-55, and gestational age 60-65 and 7 day old neonates.

Figure 6.8: MDRla rnRNA expression within the anteroventral thalamic nuclei Expression was detected at gd40. gd50 and gd60. However, expression decreased drarnatically after term and could not be detected.

Page 121: Petar - University of Toronto

Figure 6.7

40-45 50-55 60-65 7 Adults

Gestational/Neonatal age (days)

Figure 6.8

40-45 50-55 60-65 7 Adult

GestationalINeonatal age (days)

111

Page 122: Petar - University of Toronto

Figure 6.9: MDRla mRNA expression in the brain regions of fetal guinea pigs (gd50). No significant differences were observed between animals that received dexamethasone (n=9) and anirnals that received vehicle alone (n=10).

Page 123: Petar - University of Toronto

Cortex

Page 124: Petar - University of Toronto

Chapter 7

Summary and Conclusions

This study has demonstrated. for the first time, the direct effects of

endogenous and synthetic glucocorticoids on GR and MF? mRNA in fetal

hippocampal neurons. in vitro. In addition. this study examined the effects of

serotonin on expression of GR and MR mRNA in fetal hippocampal neurons, h

vitro. Examination of GR and MR mRNA was undertaken in neurons which were

derived from species with both prenatal (guinea-pig) and postnatal (mouse)

profiles of neuroendocrine development.

Studies undertaken in the mouse demonstrated that exposure to

endogenous (corticosterone) and synthetic (dexamethasone) glucocorticoids

down regulate the GR mRNA expression. Mechanisms by which this takes place

probably involve autoregulation of GR gene transcription. One possible way to

address the mechanism is to implement simultaneous GR antagonist and

glucocorticoid administration, in the same system. In this scenario, one would

expect to see no change in GR mRNA expression due to glucocorticoid

exposure. If dfierences were to be detected then one could conclude that

glucocorticoids can influence GR expression indirectly.

No differences in MR mRNA expression were detected due to either

endogenous or synthetic glucocorticoid exposure. Therefore it was concluded

that the down regulation of MR mRNA expression recorded in vivo (Levitt, et al.

1996) is probably mediated through indirect mechanisms. Serotonin exposure, in

vitro, was found to increase GR mRNA levels. This confirmed the importance of

Page 125: Petar - University of Toronto

thyroid-serotonin axis involvement in the HPA programming. Taken together,

studies undertaken in the mouse have expanded and complimented the

understanding of HPA programming in the postnatal developer model.

The guinea-pig represents a model in which extensive neuroendocrine

development occurs in ulero, a situation similar to the human. Studies in the

guinea-pig examined the direct effects of cortisol (endogenous glucocortiwid)

and dexamethasone (synthetic glucocortiwid) and serotonin on the expression of

GR and MR mRNA in fetal neurons, in vitro. This was done in a sex-specific

manner, at two gestational ages (gd40 and gd50) which are considered critical in

the guinea-pig brain development. 60th cortisol and dexamethasone exposure

down regulated GR mRNA expression in fetal neurons of both sexes. MR mRNA

expression was unaffected by glucocorticoid exposure. Serotonin treatment up

regulated GR mRNA expression in neurons of both sexes and did not affect MR

mRNA. These effects were recorded at both gestational ages considered.

Discrepancies from in vivo studies (Dean and Matthews. 1999; Dean, et al. 2000;

Matthews, et al. 2000) were explained by the presence of serotonergic system in

in vivo situation. Absence of sexual dimorphism in response ta glucocorticoids in

vitro may be explained by absence of sex hormones. Differential timing of brain

development, particularly the serotonergic system, may also be implicated in the

previously described sex differences, in vivo.

A possible functional limitation of this study was the lack of GR and MR

protein binding consideration. It would be very useful to detemine how levels of

GR and MR protein correspond to the changes in GR and MR mRNA levels. The

Page 126: Petar - University of Toronto

next step in future studies of this type would be to examine how GR and MR

protein levels change in response to glucocorticoids and serotonin. Another

functional limitation of this study is the stability of mRNA. It is possible that

observed differences in mRNA levels in this study reflect degradation of mRNA

rather than down-regulation. Examination of GWMR protein levels would also

help to resolve this issue. Another future consideration involves examination of

glucocorticoid and serotonin effects on neurons derived from fetuses at 60 days

of gestation. By this gestational time the brain of the guinea-pig displays high

neuroanatomical maturity and the process of myelination is occurring (Dobing

and Sands. 1979; Dobing and Sands. 1973). Examination of glucocorticoid

impact on hippocampal GR and MR mRNA levels at this time would offer better

insight into HPA programing.

We have also deterrnined the pattern of MDRl a mRNA expression in the

guinea pig brain. The MDRla gene was widely expressed in the developing brain

and the expression changed during development in a highly region-specific

manner. In the future studies, role of MDRla genes in the HPA programming can

be examined by using MDR -1- mouse. MDR knockout mice wuld be treated with

dexamethasone during pregnancy and their offspring allowed to reach adulthood.

Examination of wrticosteroid receptor levels in the hippocampus and

corticosterone levels in such adult animals would more definitively answer the

question of MDR gene involvement in HPA programming.

In conclusion, this study suggests that both glucocorticoids and serotonin

can direcüy influence GR mRNA and MR mRNA in developing hippocampal

Page 127: Petar - University of Toronto

neurons. Since glucocorticoids can promote maturation of the serotonergic

systern. the net effects of glucocorticoids during development on hippocampal

GR mRNA in vivo, likely depends on a balance between direct glucocorticoid

effect and indirect serotonin modulation. However. it is possible that other

aspects of brain function may be modified by prenatal glucocorticoid exposure

and that these may in tum rnodrfy corticosteroid receptor levels in the

hippocampus.

Page 128: Petar - University of Toronto

Appendix 7: Culture solutions

Minimal Essential Medium (MEM)

MEM was purchased frorn TCMP at Faculty of Medicine, University of

Toronto. 0.0259 of NaHCOJ and 3.59 of glucose was added to 500 ml of MEM.

Osmolality was adjusted with additional glucose, to 320-325 mOsM, using

osmorneter. 1 ml of rehydrated insulin (GIBCO, BRL, Insulin, Crystalinne, Bovine,

Zinc, lyophilized, cat#13007-018) was added to 500ml of MEM.

Minimal Essential Medium and NZ

MEM-N2 was prepared by adding 0.011g of sodium pyruvate (a-

Ketoproprionic acid; 2-Oxoproprionic acid; Sodium Salt; C3H303Na, FW 1 10.0;

SIGMA, P-5280, lot 68H6195). O.lg of chicken egg albumin per 100ml of MEM.

Both compounds were dissolved and MEM was filtered through 115mI

NALGENE filter, into sterile bottle. 1 ml of N2 (100X) was added per 100 ml of

MEM.

Minimal Esenfial Medium - N2 and NGF

400pL of Neural Growth Factor (NGF) solution, 40ng/pL, was added to

each ZOOml of MEM-NP. NGF was purchased from Alornone Labs.

Page 129: Petar - University of Toronto

Minimal Essential Medium containing Fetal Bovine Serum and Horse Serum

160ml of MEM was filtered through 115ml Nalgene filter, into sterile boffle.

20ml of Horse Serum (Horse Serum, heat inactivated; GIBCO: Cat. No.26050,

Lot No 1025190). and 20ml of Fetal Bovine Serurn (GIBCO: Cat. No. 26140-087,

Lot. No. 1022442) were added.

Dissection Medium

Hank's Basic Salt Solution (BSS) containing 15m HEPES was purchased

from TCMP at Faculty of Medicine, University of Toronto. 2.59 of glucose and 39

of sucrose was added to 500ml of Hank's BSS containing 15mM HEPES.

Osmolality was adjusted to 320-325 mOsm by adding glucose.

Fluorodeoxyuridine (FUDR)

0.0049 of 5-Fluoro-Zdeoxyuridine (SIGMA, F-0503) and 0.01 g of uridine

(SGMA. U-3750) were dissolved in 20ml of MEM. Solution was filtered through

1 15ml Nalgene filter.

Page 130: Petar - University of Toronto

Appendix 2:

Derivation of Treatments for Mouse Hippocarnpal Cultures

Treatment : Dexarnethasone

Stock preparation: 0.00479 of dexarnethasone was measured (Sa-Fluoro-16a-

methylprednisolone, C22H29F05, FW 392.5, SIGMA. D-1756) and dissolved in

469.59pL of 100% ethanol (concentration 25.5mM). IOpL of this solution was put

in 49990pL of PBS (concentration 5.1pM). This solution was separated into

200pL stocks. which were stored at 4'C.

Treatment: 6.5pL from stock directly to the well. This was 100nM treatment. IOpL

from stock to 90pL of MEM-FBS,HS. 6.5pL of this solution was added to each

well. This was

10nM treatment. 10pL from sock to 990pL of MEM-FBS.HS. 6.5pL of this

solution was added to each well. This was 1 nM treatment.

Calculations:

Stock: 0.01 g DW392.5/0.001L = 25.5 mM

1OpL x 25500pM = C x 50000pL; C = 5.1 VM

100 nM treatrnent: 6.5~1 x 5.1 pL = 326~ (final volume of well) x C

C = 101.5 nM

Page 131: Petar - University of Toronto

10 nM treatment: 10pL x 5.1 pM = 90pL x C; C=O.Sl pM

6.5vL x 0.51 pM = 326.5~1 x C

C = 10.15 nM

1 nM treatment: 10vL x 5.1pM = 990yL x C; C = 0.051pM

Final well volune: 50pL (cell suspension) + 250pL (volume of MEM-FBS-HS) +

20pL

(FUDR) + 6.5pL (treatment) = 326.5pL final well volume.

Similar calculations were perfonned for corticosterone and serotonin treatments.

Page 132: Petar - University of Toronto

Appendix 3: Molecular biology solutions

All of the following solutions were made with Molecular Biology Grade

reagents.

Diethyl pyrocarbonate (DEPC) treatment:

All DEPC work was camed out in a fume hood. DEPC (0.5mlllL, Omnipur,

EM Science, North Amencan associates of Merk KgaA, Darmstadt, Genany)

was added directly to each treated solution. Solution was allowed to stand in the

hood for at least 3 hours before being autoclaved.

Hybridization buffer:

Hybridization buffer was prepared a day prior to in situ hybridization (ISH).

Salmon spem was added on the day of ISH. Hybridization buffer was prepared

in a 50ml stefile polypropylene tube in following order:

25ml

1 Oml

2.5ml

0.5ml

2.5ml

1 .Oml

1 .Oml

50pL

100% deionized formamide (see below)

20X saline sodium citrate (SSC)

0.5M sodium phosphate

0.1 M sodium pyrophosphate

A00X Denhardts solution

Chlorofom-phenol extracted salmon spem DNA (1 Omglml)

Polyadenylic acid (5mgiml)

Sodium heparin (1 20mglml)

Page 133: Petar - University of Toronto

Dextran sulfate, sodium salt (Phamacia LKB)

Final volume was adjusted to 50ml with DEPC-H20. Solution was mked

vigorously, protected from light and stored at -4OC.

Deionized formamide:

Formamide (Gibco)

"AmberliteV monobed resin MB-1 (20-50 mesh; Bio-Rad

Laboratories, CA, USA)

Foramide was added to resin and the mixture was stirred at room

temperature for 2h. The mixture was filtered through a sterile filter paper and

stored at -20°C in 25ml aliquots.

Saline Sodium Citrate (20X SSC):

175.39 NaCl

88.209 Sodium citrate

50th reagents were dissolved in H20 (1 .OL) and pH was adjusted to 7.0

with HCI. Solution was then DEPC-treated and autoclaved.

Page 134: Petar - University of Toronto

Sodium Phosphate (0.5M):

Na2HP04 (0.5M)

NaH2P04 (0.5M)

The solutions were mixed and pH was adjusted to 7.0. The solution was

filtered, DEPC-treated and autoclaved.

Denhardts solution:

Polyvinylpynolidine (PVP)

Bovine semrn albumin

Ficoll

DEPC-H20

Solution was filtered and stored in aliquots (6ml) at -20°C.

Salmon Spem DNA:

Salmon sperm (10mglml) was purchased from Sigma in a ready to use

form.

Page 135: Petar - University of Toronto

Polyadenylic acid:

1 OOmg Polyadenylic acid (sodium salt)]

Ployadenylic acid was dissolved in DEPC-H20 (20ml), aliquoted (1 ml) and

stored at -20°C.

Sodium Heparin:

Freeze dtied sodium heparin was dissolved in DEPC-H20 (120mglml) and

stored in aliquots (50pL) at -20°C.

Soak Solution:

DEPC-H20

2oxssc

50% formamide

Terminal Deoxynucleotydyl Transferase (TdT):

TdT was purchased fom Gibco-BRL (FPLC pure, Calf Thymus)

5X Tailing Buffer:

Tailing buffer was supplied with the TdT (Gibco-BRL).

Page 136: Petar - University of Toronto

EDTA (0.5M):

EDTA (93.059) was dissolved in water. NaOH (log) was added and pH

was adjusted to 8.0. Final volume was adjusted to 500ml. The solution was

DEPC treated, autoclaved and stored at room temperature.

Phoshpate Buffered Saline (PBS):

8.0g/L NaCl

0.2gIL KCI

1.44gJL Na2HP04

0.24gtL NaH2P02

Paraforrnaldehyde:

Parafonaldehyde (409) was dissolved in DEPC-PBS (IL) by heating to

60°C in a fume hood. Dissolution was facilitated by NaOH addition, in a drop

wise rnanner. The pH was adjusted to 7.0 with HCI. The solution was filtered and

stored at 4°C.

Tissue fixation:

All tissues as well as cultured cells were fixed as follows:

Paraformaldehyde

PBS

PBS

5min

1 min

1 min

Page 137: Petar - University of Toronto

70% EtOH

95% EtOH

Smin

1 min

Page 138: Petar - University of Toronto

References

Ader R., Grota L.J. Adrenocortical mediation of the effects of early life experiences. Pmg Brain Res. 1973; 39: 395-406

Ajilore O.A., Sapolsky R.M. In vivo characterization of llp-hydroxysteroid dehydrogenase in rat hippocampus using glucocorticoid neuroendangement as an end point. Neuroendocnnology. 1999; 69: 138-1 44

Akner G., Wikstrom A.C., Mossberg K., Sundquist K.G., Gustafsson J-A. Morphornetric studies of the localization of the glucocorticoid receptor in marnrnalian cells and of glucocorticoid hormone-induced effects. J Histochem Cytochem. 1994; 42645-657

Almlof T., Wright A.?., Gustafsson J-A. Role of acidic and phosphorylated residues in gene activation by the glucocorticoid receptor. J Biolo Chem. 1995; 270: 17535-1 7540

Allgood V.E., Oakley R.H. Cidlowski J.A. Modulation by vitamin 86 of glucocorticoid gene mediated-expression requires translocation factors in addition to the glucocorticoid receptor. J Bi01 Chem. 1993; 268: 20870-20876

Andrele P., Niederer E., Rubas W., Hilgendorf C., Spahn-Langguth H., Wunerii- Allenspach H., Merkle HP., Langguth P. P-glycoprotein (P-gp) mediated efflux in Caco-2 cell monolayers: the influence of cultunhg conditions and drug exposure on P-gp expression levels. Journal of Pharmacetdical Sciences. 1998; 87(6): 757-762

Andrade R., Nicoll R.A. Phamacologicalyy distinct actions of serotonin on single pyramidal neurons of the rat hippocampus recorded in vitro. J Physioi. 1997; 394: 99-1 24

Anisman H., Zaharia M.D., Meaney M.J., Merali 2. Do eariy-life events pemanentiy alter behavioral and hormonal responses to stressors? int J Dev Neurosci. 1 998; 16: 149-7 64

Antoni F.A., Hypothalarnic control of adrenocorticotropin secretion: advances since the discovery of a 41-residue corticotropin releasing factor. Endocrine Reviews. 1986; 7: 351 -378

Ambrach G., Palkovits M., Szentagothai, Blood supply of the rat hypothalamus. IV. Retrochiasmic area, rnedian eminence, arcuate nucleus. Acta Morphol. Acad, Sci. Hung. 1976; 24: 93

Page 139: Petar - University of Toronto

Arina JL, Wienberger G, Cerelli TM, Glaser BL, Hendelin BL, Houseman DE, Evans RM. Cloning of human mineralocorticoid receptor complimentary DNA: structural and functional kinship with the glucocorticoid receptor. Science. 1987; 27: 268-275

Arina JL, Sirnerly RB, Swanson LW, Evans RM. The neuronal mineralocorticoid receptor as a mediator of glucocorticoid response. Neuron. 1988; 1 : 887-900

Bakker J.M., Schmidt E.D., Kroes H., Kavelaars A., Heijnen C.J., Tilders F.H.J., Van Rees €.P. Effects of short-term dexamethasone treatment dunng pregnancy on the development of the immune system and the hypothaiamo-pitutary adreanal axis in the rat. J Neuroimmunol. 1995; 63: 183-1 91

Barnberger C.M., Heinrich M., Chrousos P. Molecular deteminants of glucocorticoid receptor function and tissue sensitivity to glucocorticoids. Endocrine Reviews. 1996; 17(3): 245-261

Banker G., Goslin K. (eds.). Culturing Nerve Cells. 1991. A Bradford Book. The MIT Press, Cambridge, Massachusets, London, England. pp251-281

Barbazanges A., Piana P.V., Le Moal M., Maccati S. Matemal glucocorticoid secretion mediates long-terni effects of prenatal stress. J Neurosci. 1996; 16: 3943-3949

Barker D.J.P. Fetal and infant origins of adult disease. London, B.M.J. Books, 1991

Baxter J.D., Tyrrell J.B. In: Felig P., Baxter J.D., Broadus A.E.. Frohman L.A. (eds.), Endocrinology and Metabolism, McGraw-Hill, New York, 1987, pp. 385- 51 1

BenediMsson R., Lindsay R., Noble J., Seckl R., Edwards C.R.W. Glucocoricoid exposure in utero: A new model for adult hypertension. Lancet. 1993; 341: 339-

Bloomfield C.D., Smith K.A., Peterson B.A., Munck A. Glucocorticoid receptor adult acute lynphoblastic leukemia. Cancer Res. 1 981 ; 41 : 48574860

Bohn M.C. Glucowrtiwid induced teratolgies of the nervous system. In: Yana (ed) Neurobehavioural Teratology . 1 994; Elsevier Science ~ublishers, New York, ~~365 -387

Bohn MC., Dean D., Hussain S., Giuliano R. Development of mRNAs for glucocorticoid and mineralocorticoid receptors in the rat hippocampus. Dev Bmin &S. 1994; 77:157-162

Page 140: Petar - University of Toronto

Bourgeois S., Gruol D.J., Newby R.F., Rajah F.M. Expression of an mdr gene is associated with a new form of resistance to dexamethasone-induced apoptosis. Mol Endocrinol. 1993; 73340-851

Brann D.W., Hendry L.B., Mahesh V.B. Emerging diversities in the mechanism of action of steroid hormones. J. Steroid Biochem Mol Biol. 1 995; 52: 1 1 3-1 33

Bn'ndley D.N., Rolland Y. Possible connections between stress. diabetes, obesity, hypertension and altered lipoprotein methabolisrn that may result in artherosclerosis. Clin Sci 1989; 77: 453-461

Brink M., Humbel B.M., De Kloet E.R., Van Driel R. The unliganded glucocorticoid receptor is localized in the nucleus, not in the cytoplasm. Endocrinolgy. 1992; 1 30: 3575-3581

Brock T.D., Madigan M.T., Martinko J.M. Parker J. Biology of Microorganisms. seventh edition. 1994; Prentice Hall, Engelwood Cliffs, New Jersey 07632; p35

Broody M.J., Haywood J.R., Touw K.B. Neural rnechanisms in hypertension. Annu Rev. Physiol. 1980; 42: 441 -453

Bumstein K.L., Bellingham DL., Jewel C.M., Powel-Olive F.E. Cidlowski J.A. Autoreg u lation of glucocorticoid receptor gene expression. Steroids. 1 991 ; 56:25-

Cato ACB, Mink S, Hartig E. Gene activation mineralocorticoid receptors: the role of the Farman N, Lombes M, Rafestin-Oblin M-E

is mediated by glucocorticoid and steroid ligand. In: Bonvalet JP, (eds). Aldosterone: Fundamental

Aspects. Libbey Eurotext, Pans and London. 1 991 : pp23-32

Cavanaugh A.H., Sirnons S.S.J. Factor-assisted DNA binding as a possible general mechanism for steroid receptors. J Steroid Biochem Mol Biol. 1994; 48 ~433-446

Chen C.J., Chin J.E., Ueda K., Clah D.P.. Pastan I., Gottesman M.M., Roninson I.B. Interanal duplication and homolgy with bacterial transport proteins in the mdrl a (P-glywprotein) gene frorn rnulti-drug resistant human cells. Cell. 1986; 47~381-389

Cidlowski J.A., Cidlowski N.B. Regulation of glucocorticoid receptor by glucocorticoids in cultured HeLa cells. Endocnnology. 1981 ; 109: 1975-1 982

Cintra A.. Slofrini V., Bunnemann B .. Okret S., Bartolotti F., Gustafsson J-A., Fuxe K. Prenatal development of glucowrticoid receptor gene expression and immunoreactivity in the rat brain and pituitary gland: a combined in situ

Page 141: Petar - University of Toronto

hybridization and immunocytochemical studies. Neuroendocnnology. 1993; 57:Il33-1147

Clement A.D. The incidence of attention deficit-hyperactivity disorder in children whose mothers experienced extreme psychological stress. Georgia Educational Research. 1992; 91 (1 &2):1-14

Croop LM., Raymond H., Haber D., Devault A., Arceci R.J., Gros P., Housman D.E. The three rnouse rnultidrug resistence (mdr) genes are expressed in a tissue specific manner in normal mause tissues. Moi Ceii Biol. 1989; 9:1346- 1350

Dahmer M.K., Tienwngroj W., Pratt WB. Purification and preliminary charactenzation of a macromolecular inhibitor of glucocorticoid receptor binding to the DNA. J Biol Chem. 1985; 260: 7705-7715

Dallman M.F., Akana S.F., Levin N., Walker GD., Bradbury M.J., Suemary S., Scribner K.S. Corticosteroids and the control of function in the hypothalamo- pituitary -adrenal (HPA) axis. Ann NY Acad Sci. 1994; 746: 22-32

Danielsen M, Northrop JP, Ringold GM. The mouse glucocorticoid receptor: mapping of functional domains by cloning, sequencing and expression of wild- type and mutant receptor proteins. EMBO J. 1986; 5(10): 251 3-2522

Dean F., Matthews S.G. Matemal dexamethasone treatment in late gestation alters glucocorticoid and mineralocorticoid receptor mRNA in the fetal guinea pig brain. Brain Res. 1999; 846: 253-259

Dean F., Yu C., Matthews S.G. Matemal glucocorticoid treatment in late gestation programs hypothalmo-pituitary adrenal function in guinea pig offspring. Endocrine Society 1999, San Diego, Abstract P 2:397

De Kloet R.E., Vreugdenhil E., Oitzl S.M., Joels M. Brain corticosteroid receptor balance in health and disease. Endocrine reviews 1998; 19(3): 269-301

De Kloet E.R. Brain corticosteroid receptor balance and homeostatic control. Frontiers Neuroendocrinol. 1991 ; 12: 164

De Kloet ER., Wallach G., McEwen B.S. Differences in corticosterone and dexamethasone binding to rat brain and pituitary. Endocrinology. 1975; 96: 598- 609

Diaz R., Brown R.W., Seckl J.R. Distinct ontogeny of glucocorticoid and mineralocorticoid receptor and 11 p-hydroxysteroid dehydrogenase types I and II mRNAs in the fetal rat brain suggest a complex control of glucocorticoid actions. J Neurosci. 1998; 1 8:25?Oœ2580

Page 142: Petar - University of Toronto

Dinan TJ. Serotonin and the regulation of hypothalamic-pituitary-adrenal axis function. Life Sciences. 1996; 58(20): 1 683-1 694

Dobbing J., Sands J. Quantitative growth and development of human brain. Arch Dis Chiid. 1973; 48: 757-767

Dobbing J., Sands J. Comparative aspects of the brain growth spurt. Early Hum Dev. 1979; 3: 79-83

Dobbing J., Sands J. Growth and development of the brain and spinal chord of the guinea pig. Brain Res. IWO; 17: 1 15-1 23

Dodic M., Peers A., Wintour E.M., Coghlan J.P. Can excess glucocorticoids, in utero, predispose to cardiovascular and metabolic disease in middle age? Trends Endocnnol Metab. 1999; 10: 86-91

Dodt C., Kem W., Fehm H.L., Born J. Antimineralocorticoid canrenoate enhances secretoty activity of the hypothalamic-pituitary-adrenal (HPA) axis in humans. Neuroendocrinology. 1993; 58: 570-574

Dunn J.D., Orr S.E. Differential plasma corticosterone responses to hippocamapal stimulation. Exp. Brain Res. 1984; 54: 1-6

Eipper B.A., Mains R.E. Structure and biosynthesis of proadrenocorticotropin/endorphn and related peptides. Endocrine reviews. 1980; 9: 135-1 58

Elliot E.M., Sapolsky R.M. Corticosterone enhances kainic acid-induced calcium elevation incultured hippocampal neurons. J. Neurochem. 1992; 59: 1033-1 040

Elliot E.M., Sapolsky R.M. Corticosterone impairs hippocarnpal neuronal calcium regulation -possible mediating mechanisms. Brain Res. 1993; 602: 84-90

Elvidge H. Production of dated pregnant guinea pigs without post-partum mating. J. lnst Anim. Tech. 1972; 23: 1 1-1 17. Abstract.

Evans RM, Ariza JL. A molecular framework for the actions of glucocorticoid homones in the nervous system. Neuron. 1989; 2: i lO5- l l l2

Eytan GD.. Borgnia M.J. Regev R., Assarf Y.G., 1994.J. Biol. Chem. 269: 26058-26065

Feldman S., Conforti N. Participation of the dorsal hippocampus In the glucocorticoid feedback effects on adrenocortical activity. Hom Res. 1980; 30: 52

Page 143: Petar - University of Toronto

Fench N.P. Hagan R., Evans S.F.. Godfrey M., Newnham J.P. Repeated antenatal corticosteroids. Amencan Journal of Obstetncs and Gynecology. 1999; 180: 114-121

Field T., Sandberg D., Queetel T.A., Garcia R., Rosario M. Effects of ultrasound feedback on pregnancy, anxiety, fetal activity, and neonatal outcorne. Obstetncs and Gyneacology. 1985; 66: 525-528

Gerlach J.L., McEwen B.S. Rat brain binds adrenal steroid hormone: radioautography of hippocampus. Science. 1972; 175:1133-1136

Gomez-Sanchez E.P. Central hypertensive effects of aldosterone. Front Neuroendocrinol. 1997; 18: 440-462

Gould E. The effects of adrenal steroids and excitatory input on neuronal birth and survival. Ann NY Acad Sci 1994; 743: 73-94

Gould E., Wooley CS., McEwen B.S. Adrenal steroids regulate postnatal development of the rat dentate gynis: 1. Effects of glucocorticoids on cell death. J Comp Neurol. 1 991 ; 31 3: 479-485

Gros P., Raymond M., Bell J., Housman D. Cloning and characterization of a second member of the mouse mdr gene family. Mol Cell Biol. 1988; 8: 21 10-2778

Herman J.P., Prewitt C.M., Cullinan W.E. Neuronal circuit regulation of the hypothalamo-pitu itary-ad renocortical stress axis. Crit Rev Neurobiol. 1 996; 1 0: 371 -394

Herman J.P., Schafer M.H., Young E.A., Thompson R., Doug J., Akil H., Wastson S.J. Evidence for hippocampal regulation of neuroendocrine neurons of the hypothalamo-pituitary-adrenocortical axis. J Neurosci. 1989; 9:3072

Hessen W., Karten Y.J.G., Van de Witte S.J., Joels M. Serotonin and carbachol induced supression of synaptic excitability in rat CA1 hippocampal area: effects of corticosteroid activation. J Neuroendocrinol. 1998; 8: 433-438

Hu LM., Bodwell J., Hu J-M., Orti E., Munck A. Glucocorticoid receptors in ATP- depleted cells. J Biol Chem. 1994; 269: 6571 -6577

Hua S.Y., Chen Y.Z. Membrane receptor-mediated electrophysiological effects of glucuwrtiwid on mammalian neurons. Endocnnology. 1989; 124: 687-691

Page 144: Petar - University of Toronto

Jacobson L., Sapolsky R.M. The role of hippocampus in the feedback regulation of the hypothalamic-pituitary-adrenocorticai axis. Endocrine Reviews. 1991 ; 12(2): 1 1 8-1 34

Joels M., De Kloet E.R. Mineralocorticoid receptor-mediated changes in membrane properties of rat CA1 pyramidal neurons in vitro. Proc Natl Acad Sci USA. 1990; 87: 4495-4498

Joels M., Femhout B. Decreasesd population spike in CA1 hippocampal area of adrenalectornized rats afier repeated synapüc stimulation. J Neuroendocfinol. 1993; 5: 537-543

Joels M., De kloet E.R. Coordinative mineralocorticoid and glucocorticoid receptor-mediated control of responses to serotonin in rat hippocampus. Neuroendoctfnology. 1992; 55: 344-350

Karst H., Wadman W.J., Joels M. Corticosteroid receptor-dependent modulation of calcium currents in rat hippocampal CA1 neurons. 1994; Brain Res. 649: 234- 242

Keightley M-C.. Fuller P.J. Unique sequences in the guinea-pig glucocorticoid receptor induce constitutive transactivation and decrease steroid sensitivity. Mol. Endocnnology. 1 994; 8: 431 -439

Kerr D.S.. Campbell L.W., Thibault O., Landfield P.W. Hippocampal glucocorticoid receptor activation enhances voltage-dependent Ca2+ conductances: relevance to brain aging. Proc Nat1 Acad Sci USA. 1992; 89: 8527-8531

Kralli A., Bohen S.P.. Yamamoto K.R. LEMI, the ATP-binding cassette transporter, selectively modulates the biological potency of steroid hormones. Proc Natl Acad Sci USA. 1995; 92:47Ol=4iO5

Krozowski Z.S., Funder J.W. Renal mineralocorticoid receptors and hippocampal corticosterone binding species have identical intrinsic steroid specificity. Proc. Nat! Acad Sci USA. 1983: 80: 6065-6060

Langley-Evans S.C. Matemal carbenoxolone treatrnent loweres birth weight and induces hypertension in the offspring of rats fed a protein-replete diet. Clin Sci. i 997; 423-429

Lankas G.R., Wise L.D., Cartwright M.E., Pippert T., Umben hauer D.R. Placental P-glywprotein deficiency enhances susceptibility tu chemically induced birth defects in mice. Reproductive Toxicoiogy. 1 998; 1 2(4): 457-463

Page 145: Petar - University of Toronto

Levine S. Maternal and environmenta! influences on the adrenocortical response to stress in weaning rats. Science. 1957; 156: 258-260

Levitt N.S., Lindsay R.S., Holmes M.C., Seckl J.R. Dexamethasone in the last week of pregnancy attenuates hippocarnpal glucocorticoid receptor gene expression and elevates blood pressure in the adult offspring rat. Neuroendocrinology. 1996; 64: 412-41 8

Liu D, Diorio J, Tannenbaum B, Caldji C, Francis D. Freedman A, Sharma S, Pearson 0, Plotsky PM, Meaney MJ. Maternal care, hippocarnpal glucocorticoid receptors, and hypothalamic-pituitary-adrenal response to stress. Science. 1997; 277: 1659-1 662

Loe D.W ., Sharom F. J., 1994. Biochim. Biophys. Acta. 1 190: 72-84

Loo T.W., Clarke D.M. Inhibition of oxidative cross-linking between engineered cysteine residues at positions 332 in predicted transrnernbrane segments (TM) 6 and 975 in predicted TM12 of human P-glycoprotein by drug substrates. J. Biol. Chem. 1996; 271 : 27482-27484

Lupien S.J., McEwen B.S. The acute effects of corticosteroids on cognition: integration of animal and human mode1 studies. Brain Res Brain Res Rev. 1997; 24: 1-27

Nakamura Y., Ikeda S., Furukawa T., Surnizawa T., Tani A., Akiyama S., Nagata Y. Function of P-giycoprotein expressed in placenta and mole. Biochem and Biophys. Res Commun. 1997; 235: €349-53

MacLusky N.J., Clark AS., Naftolin F. Goldman-Rakic P.S. Estrogen formation in the rnammalian brain: possible role of aromatase in sexual differentiation of the hippocampus and cortex. Steroids. 1987; 50: 459-474

Madison D.V., Nicoll R.A. Actions of noradrenaline recorded intracellularly in rat hippocampal pyramidal neurons, in vitro. J Physiol. 1986; 372: 221 -244

Magistretti J., De Curtid M., Vescovi A., Galli R., Gritti A. Long-term survival of cortical neurones from adult guinea-pig maintained in lowdensrty cultures. Neumport. 1996; 7: 1559-1 564

Mason S.A., Housley P.R. Sitedirected mutagenesis of the phosphorylation sites in the rnouse glucocorticoid receptür. J Biol Chem. 1995; 268: 21 501 -24 504

Mason J. W . The CNS regulation of ACTH secretion, In: The reticulat formation of the brain (H. H Casper, L.D. Proctor, R.S. Knighton, W.C. Noshay, R.T Costello, eds.) Little Brown. Co., Boston (1958): 645

Page 146: Petar - University of Toronto

Matthews S.G. Antenatal glucocorticoids and programming of the developing CNS. Pediatric Research. 2000; 47(3): 291 -300

Matthews S.G. Li A., Liu L. Repeated glucocorticoid treatment of pregnant guinea-pigs programs pituitary-adrenal function in aduR offspring in a highly sex- specific manner. Endocrine Society, Toronto 2000. Abstract 877

Matthews S.G. Dynamic changes in glucocorticoid and rnineralocorücoid receptor mRNA in the developing guinea pig brain. Developmental Brain Res. 1998; 52: 498

Matthews S.G., Yang K., Challis J.R.G. Changes in glucocorticoid receptor mRNA in the developing ovine pituitary and the effects of exogenous cortisol. Joumal of Endocrinology. 1 995; 144: 483490

Matthews S. G.. Challis J.R.G. Regulation of CRH and AVP mRNA in the developing ovine hypothalamus: effects of stress and glucocorticoids. Amencan Journal of Physiology: Endocrinology and Metabolism. 1 995; 268: E l 096-El 107

McEwen B.S., Weiss J.M., Schwartz L.S. Selective retention of corticosterone by limbic structures of the brain. Nature. 1968; 220: 91 1-97 2

McEwen B.S., De Kloet E.R., Rostene W. Adrenal steroid receptors and actions in the nervous system. Physioi. Rev. 1986; 66: 1 121 -1 188

McKenna N.J., Xu J., Nawaz Z., Tsai M.J.. O'Malley B.W. Nuclear receptor coactivators: multiple enzymes, multiple coprnlexes, multiple functions. J Steroid Biochern Mol Biol. 1999; 69(1-6): 3-1 2

Meaney M.J., Sapolsky R.M., McEwen B.S. The development of the glucocorticoid receptor system in the rat limbic brain: I 1. An autoradiographic study. Dev Brain Res. 1 985; 18: 165-1 68

Meaney MJ, Diorio J, Francis D, Larocque S, O'Donnell D, Smythe JW, Shamia S, Tannenbaum B. Environmental regulation of the developrnent of glucocorticoid receptor systems in the rat forebrain: the role of serotonin. A m N Y Acad Sci. 1994; 746: 260-274

Meaney MJ, Aitken DH, Sapolsky RM. Thyroid hormones influence the development of hippocampal glucocorticoid receptors in the rat: A mechanisrn for the effects of postnatal handling on the developrnent of the adrenocortical stress response. Neuroendocrinology. 1 987; 45: 278-283

Meibach R.C., Siegel A. Efferent connections of the hippocampal formation in the rat. Brain Res. 1977; 124:197

Page 147: Petar - University of Toronto

Meijer O.C., De Lange E.C.M., Breimer D.D.. De Boer A.G.. Workel J.O., De Kloet E.R. Penetration of dexamethasone into brain glucocorticoid target is enhanced in mdrl a P-glycoprotein knockout mouse. Endoc~fnology. 1998; 139(4): 17898-1 793

Michelson D., chrousos G.P., Gold P.W. Type 1 glucocorticoid receptor blocade does not afect baseline or ovine corticotropin releasing hormone stirnulated adrenocorticotropic hormone and cortisol secretion. Neuroimmunomodulation. 1994; 1 : 274-277

Mitchel JB, Rowe W. Boksa P, Meaney MJ. Serotonin regulates type II corticosteroid binding in hippocampal cell cultures. The Journal of Neuroscience. 1990; 1 O(6): 1745-1 752

Mitchell JB, Iny LJ, Meaney MJ. The role of serotonin in the development and environmental regulation of type II corticosteroid receptor binding in rat hippocampus. Developmental Brain Research. 1 990; 55: 231 -235

Motta M., Piva F., Martini L. The role of 'short' feedback mechanisms in the regulation of adrenocorîicotropin secretion, in De Wied and WeijnenPituitary, adrenal and the brain. Prog. Brain Res. 1970; 32: 25-32

Munck A.. Guyre P.M., Holbook N.J. Physiological functions of glucocorticoids in stress and their relation to phanacological actions. Endocrine Reviews. 1984; 5: 25-44

Munck A., Naray-Fejes-Toth A. The ups and downs of glucocorticoid physiology. Premissive and supressive effects revisited. Mol Ce11 Endocnnol. 1992; 90: Cl- C4

Munck A., Naray-FejesToth A. Glucocorticoids and stress: premissive and supressive actions. Ann NY Acad Sci. 1994; 746:115-120

Muneoka K., Mikuni M., Ogawa T., Kitera K., Kamei K., Takigawa M.. Takahashi K. Prenatal dexamethasone alters brain monoamine metabolism and adrenocortical response in rat offspring. Am J Physiol. 1997; 273: RI 669-RI 675

OYKeefe J.R., Handa R.J. Transient elevation of estrogen receptors in the neonatal rat hippocampus. Dev. Brain. Res. 1990: 57(1): 1 19-127

Nauta W.J.H. An experimental study of the fomix systern in the rat. J. Comp. Neurol. 1956; 104: 247

Page 148: Petar - University of Toronto

NIH Consensus Development Panel on the effects of corticosteroids for fetal maturation on perinatal outcomes. JAMA. 1995; 273: 41 3-41 8

Oppenheimer J.H., Schwark H.L. Molecular basis of thyroid hormone-dependent brain development. Endocr Rev. 1997; 18: 462-475

Plotsky P.M., Cunningham JR E.T.. Widmaier E.P. Catecholaminergic modulation of corticotropin-releasing factor and adrenocorticotropin secretion. Endocrine Reviews. 1 989; 1 O(4); 437458

Plotsky P. M. Regulation of hypophysiotropic factors mediating ACTH secretion. In: The hypothalamic pituifary adrenal axis revisited. (W. Ganong, M. Dallman, J. Roberts Eds.). New York Academy of Science, New York. 1987; 512: 205

Plotsky PM, Meaney MJ. Early postnatal experience alters hypothalamic corticotrophin-releasing factor (CRF) mRNA, median eminence CRF content and stress-induced release in adult rats. Brain Research Molecular Brain Research. 1993; 18: 195-200

Ratka A., Sutanto W., Bloemers M., De Kloet E.R. On the role of brain mineralocortirnid (type 1) and glucocorticoid (type II) receptors in neuroendocrine regulation. Neuroendocnnology. 1989; 50: 1 17-1 23

Reul J.M.H.M., De Kloet ER. Two receptor systems for corticosterone in rat brain: microdistribution and differential occupation. Endocnnology. 1 985; 1 1 7: 2502-251 1

Robertson N.M., Bodine P.V., Hsu T.C., Alnemri ES., Litwack G. Modulator inhibits nuclear translocation of the glucocorticoid receptor in the hurnan leukernic cell line Cem C-7. Cancer Res. 1995; 55548-556

Rosenberg M.F., Callaghan R., Ford R.C., Higgins C.F. Structure of the multidrug resistance P-glycoprotinein to 2.5nm resloution determined by electron microscopy and image analysis. Journal of Biologicai Chemistry. 1 997; 272: 1 O685

Rosenfeld P., Van Eekelen J.A.M., Levine S., De Kloet E.R. Ontogeny of corticosteroid receptor system in the brain. Ceil Mol Neurobiol. 1993; 13:295-319

Rubin R.T.. Mandel A.J., Crandall P.H. Corücosteroid responses to limbic stimulation in man: localization of stimulus sites. Science. 1966; 153: 767

Rue& S., Gros P. Functional expession of P-glycoproteins in secretory vesicles. J Bi01 Chem. 1994; 269:12277-12228 Sarkadi B.. Muller M., Homolya L., Hollo Z., Seprodi J., Germann U.A., Gottesman M.M., Price E.M., Boucher R.C. 1994. FASEB J. 8: 766-770

Page 149: Petar - University of Toronto

Rupprecht R, Anna JL, Spengler O, Reul JMHM, Evans RM, Holsboer F, Damrn K. Transactivation and synergistic properties of the mineralocorticoid receptor: relationship to the glucocorticoid receptor. Molecular Endocrinology. 1993; 7: 597-603

Sanchez E.R.. Hu J-L., Zhong S. Shen P., Greene M.J., Housleey P.R. Potentiation of glucocorticoid receptor-mediated gene expression by heat and chernical shock. Mol Endocniiol. 1994; 8:408-421

Sapolsky R.M., Krey L.C., McEwen B.S. Glucocorticoid-sensitive hippocarnpal neurons are involved in terrninating the adrenocortical stress response. Proc. Natl. Acad. Sci. USA. 1984; 81: 6174

Sapolsky R.M., Meaney M.J. Maturation of adrenocortical stress response: Neuroendocrine control mechanisms and the stress hyporesponsive period. Brain Res. Rev. 1 986; 1 1 35-76

Schmidt T.J., Miller-Diener A., Webb M.L., Litwack G. Thermal activation of the purified rat hepatic glucocorticoid receptor. Evidence for a two-ster mechanism. J. Biol. Chem. 1 985; 260: 1 6255-1 6262

Schinkel A.H. The physiological function of drug-transporting P-glycoproteins. Cancer Biology. 1 997; 8: 161 -1 70

Schinkel A.H., Wagenaar E., Mol C.A.A.M., Van Deemter L. P-glycoprotein in the blood-brain barrier of mice influences the brain penetration and phanacological activity of many drugs. J. Clin. Invest. 1996; 97:2517-2525

Schinkel A.H., Wagenaar E., van Deemter L., Mol C.A.A.M., Borst P. Absence of the mdrl a P-glycoprotein in mice affects tissue distribution and pharmacokinetics of dexamethasone, digoxin and cyclosporin A. J Clin. Invest. 1995; 96: 1698- 1705

Schinkel A.H., Smit J.J.M., van Tellingen o., Beijnen J.H., Wagenaar E., van Deemter L., Mol C.A.A.M., van der Valk M.A., Robanus-Maandrag E.C., te Riele H.P.J., Bems A.J. M.. Borst P. Disruption of the mouse mdrla P-glycoprotein gene leads to a deficiency in the blood-brain barrier and to increased sensitivity to dnigs. 1994; Cell. 77: 491 -502

Seckl J.R. 1 *i pHydroxysteroid dehydrogenase in the brain: a novel regulator of glucawrtiwid action? Front Neuroendocrinol. 1997; 18: 49-99

Seyle H. The story of the adaptation syndrome. Acta Inc., Medical Publishers, Montreal, Canada

Page 150: Petar - University of Toronto

Shama R.C., houe S., Roitelman J., Schimke R.T., Simoni R.D. 1992. J. Biol. Chem. 267: 731-5734

Sharom F.J. The P-glycoprotein efflux pump: how does 1 transport drugs? J. Membrane Biol. 1997; 160: 161 -1 75

Sharom F.J. 1995. J. Bioenerg. Biornembr. 27: 15-22

Sharom F.J., Yu X., DiDiodato G., Chu J.W.K. 1996. Biochem J. 320: 421-428

Silva CM, Powell-Oliver F, Jewell CM, Sar Ml Allgood VE, Cidlowski JA. Regulation of the human glucocorticoid receptor by long-terrn and chronic treatment with glucocorticoid. Steroids. 1994; 59: 436-442

Slotkin TA, Bames GA, McCook EC. Seidler FJ. Programming of brain stem serotonin transporter development by prenatal glucocorticoids. Developmental Bmin Research. 1 996; 93: 155-1 61

Slotkin T.A., Lappi S.E., McCook E.C., Tayyeb M.I., Eylers J.P., Seidler F.J. Glucocorticoids and the development of neuronal function: effects of prenatal dexamethasone exposu re on central no rad renerg ic activity. Bi01 Neonate. 1 992; 61 : 326-336

Slotkin T.A., Lappi S.E., Tayyeb M. I., Seidler F. J. Dose-dependent glucucorticoid effects on noradrenergic synaptogenesis in rat brain: ontogeny of [3H]desmethyllimipramine binding sites after fetal exposure to dexamethasone. Res Commun Chem Pathol Pharmacol. 1 99 1 ; 73:3-19

Smith D.F., Toft BO. Steroid receptors and their associated proteins. Moi Endocnnol. 1993; 7: 4-1 1

Storm J.F. Potassium currents in hippocampal pyramidal cells. Prog Brain Res. 1990; 83: 161-187

Sutanto W., De kloet E.R. Species-specificity of cortiwsteroid receptors in hamster and rat brains. Endocnnology. 1987; 121 : 1405

Svec F., Rudis M. Glucocorticoids regulate the glucocorticoid receptor in the AtT- 20 celi. J. Biol. Chem. 1981 ; 256: 5984-5987

Swanson LW., Cowan M.W. An autoradiographic study of the organization of the efferent connections of the hippocampus formation in the rat. J. Comp. Neurol. 1 977; l72:49

Swanson L.W., Sawchenko P.E., Berod A., Hartman B.K., Helle K.B., Vanorden D.E. An irnmunohistochemical study of the organization of catecholaminergic

Page 151: Petar - University of Toronto

cells and teminal fields in the paraventricular and supraoptic nuciei of the hypothalamus. The journal of Comparative Neurology. 1 981 ; 1 96:271-285

Takahashi L.K. Prenatal stress: consequences of glucocorticoids on hippocampal development and function. Int J dev Neurosci. 1 998; l6:199-207

Thiebaut F., Tsuruo T., Hamada H., Gottesman M.M., Pastan I., Willinghham M.C. Cellular localization of the rnultidrug resistance gene product in normal human tissues. Proc. Nat/ Acad Sci USA. 1 987; 84:7735-7738

Truss M., Bartcsh J., Schelbert A., Hache RJG., Beato M. Hormone induces binding of receptors and transcription factors to a rearranged nucleosome on the MMTV promoter in vivo. € M O J. 1995. 14: 1737-1751

Ueda K., Okarnura N., Hirai M., Tanigawara Y., Saeki T., Kioka N., Komano T., Hori R. Human P-glycoprotein transports cortisol, aldosterone, and dexamethasone but not progesterone. J Bi01 Chem. 267: 24248-24252

Uno H., Eiself S., Sakai A., Shelton S., Baker E., DeJesus O., Holden J. Neurotoxicity of glucocorticoids in the primate brain. Hormone and Behavior. 1994; 28: 336-348

Uno H., Lohmiller L., Thieme C., Kemnitz J.W., Engle M.J., Roecker E.B., Farrel P. M. Brain damage induced by prenatal exposure to dexamethasone in fetal rhesus macaques. Developmental Brain Research. 1990; 53: 1 57-1 67

Van Eekelen J.A.M., Bohn M.C., De Kloet E.R. Post natal ontogeny of mineralowrticoid and glucocorticoid receptor gene expression in the regions of the rat tel- and diencephalon. Dev Brain Res. 1991 ; 61 :33-44

Van Haarst A.D., Oitzl M.S., De kloet E.R. Facilitation of feedback inhibition through blockade of glucocorticoid receptor in the hippocampus. Neumchem Res. 1997; 22: 1323-1 328

Van Kalken C.K., Giaccone G., van der Valk P., Kuiper C.M., Hadisaputro M.M.N., Meijer C. J LM., Piendo H.M. Multidrug resistence gene (P-glycoprotein) expression in the human fetus. Amencan Journal of Pathology. 1992; 141 (5): 1063-1 072

Veldhius HD, Van Koppen C, Van lttersum M, De De Kloet ER. Specificity of the adrenal steroid receptor system in rat hippocampus. Endocnnolgy. 1982; Il O: 2044-2049

Vivanco M.D., Johnson R.. Galante P.E., Hanahan D., Yamamoto KR. A transition in transcriptional activation by the glucocorticoid and retinoic acid

Page 152: Petar - University of Toronto

receptors at the tumor stage of dermal fibrosarcoma developrnent. EMBO J. 1995; 14~2217-2228

Walker BR., Williams B.C. Corticosteroids and vascular tone: mapping the messenger maze. Clin. Sci. 1992; 55:376-385

Webster J.C., Cidlowski J.A. Downregulation of the glucocorticoid receptor: a mechanism for physiological adaptation to hormones. Ann NY Acad Sei. 1994; 746: 21 6-220

Weinstock M. Does prenatal stress impair coping and regulation of hypothalamic- pituitary-adrenal axis. Neurosci Behav. 1997; 21 :1-10

Whitnall M. H. Regulation of the hypothalarnic corticotropin -releasing hormone neurosecretory system. Progress in Neurobiology. 1993; 40(5): 573-629

Wilson M., Greer S.E., Greer M.A., Roberts L. Hippocampal inhibition of pituitary-adrenocortical function in female rats. Brain Res. 1980; 197:433