Effects ofEstrogen in the Basolateral Amygdala ofthe Rat Brain by James Arthur Andrews Submitted in Partial Fulfillment ofthe Requirements for the Degree of Master ofScience in the Biological Sciences Program Youngstown State University June, 2000 Effects ofEstrogen in the Basolateral Amygdala ofthe Rat Brain by James Arthur Andrews I hereby release this thesis to the public. I understand this thesis will be housed at the Circulation Desk ofthe University Library and will be available for public access. I also authorize the University or other individuals to make copies ofthis thesis as needed for scholarly research. Signature: Approvals: ~~, 1L~ Mark D. Womble, Thesis Advisor alker, Committee Member ~/ -/ ~ it:.-) .c>t.-<-t~ " ~ ~u"- Robert . LeiPhzr, Committee Member Peter J. Nasvinsky, Dean of Date ,61160 Date ~_ .. JI/OC Date ~L!fttr? Date Date Acknowledgements Indeed, I am grateful and indebted to all ofthose who contributed to my works published here. Including all ofthe beneficiaries ofmy gratitude in one document is impossible, so please forgive me ifI do not list your name. My friends and family have performed harder than I at times. I love you all. The amount ofhelp they contributed is beyond quantification. Additionally, thanks to those who lent me shelter when I had none. Above all, thanks and praise to the Creator. Without faith, I doubt that I would have finished this project. You gave the determination, knowledge, and wit to deal with the relative enormosities encountered in this project. When I needed motivation, you were there for me. My Grandfather, Arthur E. Verner is the first non-omnipotent being to which I must give thanks (not that he is too far from it). When I was unable to continue, I thought ofall ofthe times he went out ofhis way to make sure that I was well in difficult times. In times without food, without water, or without money...he has always come through. Most importantly, his statement: "Ifyou're gonna start something, finish it!" echoed in my head, and drove me to research, read, and write to create the thesis contained within the following pages. 111 Abstract Clinical evidence has shown estrogen may delay the onset of Alzheimer's disease and protect against neuronal damage associated with stroke. Intracellular recordings (current-clamp) were made to characterize the effects ofestrogen in the basolateral amygdala (BLA) ofthe rat brain. Excitatory postsynaptic potentials (EPSPs) were elicited by stimulation of afferents in the external capsule. Estrogen was found to decrease EPSP amplitude in a rapid (20-30 min) fashion. Similarly, reduction of spontaneous synaptic activity occurred upon estrogen treatment. EPSP amplitudes returned to normal within 20 minutes ofestrogen washout. 4 hydroxy tamoxifen (4-0HT), and estrogen receptor antagonist, prevented the estrogen-induced decrease in EPSP amplitude, suggesting dependence on an estrogen receptor. Estrogen treatment had no effect on neuronal input resistance, accommodation response, resting membrane potential, or action potential firing frequency. Preliminary data showed no change in inhibitory postsynaptic potential (IPSP) amplitude, suggesting estrogen might act on the presynaptic cell. These findings imply that estrogen may be protecting neurons from excitotoxic injury associated with stroke through the modulation ofglutamate release. IV Table ofContents Title Page i Signature and Release .. Acknowledgements iii Abstract iv Table ofContents v Table ofFigures .. Introduction 1 Alzheimer's disease 1 Treatment ofAlzheimer's Disease 5 Heredity ofAlzheimer's Disease 7 Estrogen 8 The Genomic Pathway ofEstrogen Action 12 The Estrogen Receptor (ER) 13 Antagonism ofthe ER 15 Non-Genomic Mechanism ofEstrogen Action 17 The Basal Lateral Amygdala (BLA) 20 v Materials and Methods 25 Results 29 Discussion 67 References 82 VI Table ofFigures Figure 1. The Pathway ofestrogen synthesis 11 Figure 2. Stimulating/Recording apparatus 24 Figure 3. Effect ofestrogen on EPSP amplitude 36 Figure 4. Effects ofestrogen on cellular recordings ofEPSPs from BLA neurons superimposed 37 Figure 4a. EPSPs in control saline and estrogen 37 Figure 4b. BLA EPSP responses under perfusion ofestrogen and estrogen washout superimposed 38 Figure 5. Indiyidual cellular recordings ofEPSPs from a BLA neuron presynaptically stimulated via EC 39 Figure Sa. EPSP recordings made in the presence ofcontrol ACSF 39 Figure 5b. EPSP recordings made in the presence of estrogen 40 Figure.5c. EPSP recordings after switching the flow ofbath solution from estrogen to normal ACSF 41 Figure. 6. Individual cellular recordings oftrain stimulations from BLA neurons superimposed 42 Figure. 6a. Train stimulations in control ACSF and ACSF containing estrogen 42 VB Figure.6b. Superimposed train stimulations in ACSF containing estrogen and estrogen washout. .. """""" """"" "" "." """ """ " " 43 Figure 7. Individual cellular recordings from a BLA neuron train stimulated via EC. " """" """"" """""" " " ".. " ".""'"~ 44 Figure 7a. Train stimulation recordings ofmade from a BLA neuron under train stimulation via EC in control ACSFII"'"''''''''''''''''''''''''''''''''''''''''44 Figure 7b. Train stimulation recordings ofmade from a BLA neuron under train stimulation via EC in ACSF containing estrogen..""""""".""."" 45 Figure 7c. Train stimulation recordings ofmade from a BLA neuron under train stimulation via EC after estrogen washout. "".""."""."""".".""." 46 Figure 8. Effect ofthe alcohol vehicle on EPSP amplitude. """""""".""" 47 Figure 9. Individual cellular recordings ofEPSPs from BLA neurons with the effects ofcontrol, ethanol, and estrogen superimposed. """""""."" 48 Figure 9a. Single BLA neuron EPSP responses from a single cell perfused with control ACSF and ACSF containing alcohol superimposed. """". 48 Figure 9b. Single BLA neuron EPSP responses from a single cell perfused with estrogen and ethanol superimposed. "."""".""."".""".""""""""" 49 Figure 1Q. Effect ofestrogen on intracellular recordings ofIPSPs from BLA neurons superimposed.."""""" """""""." "".""."" " "" 50 V111 Figure lOa. BLA IPSP responses from a single cell perfused with control ACSF and wash superimposed. """"""",,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,"",,,,,,, 50 Figure 1Ob. BLA neuron IPSP responses from a single cell perfused with estrogen and estrogen washout superimposed. """"""""""""""""""".51 Figure II. Individual IPSP recordings from a BLA neuron presynaptically stimulated via EC.."""".. ".. "."""""""."""""...""."".""...".. """",""", 52 Figure 11 a. Recordings ofIPSPs made from a BLA cell under presynaptic stimulation via EC in control ACSF. """"""""""""""".""""""""""". 52 Figure lIb. IPSPs recordings from a single cell perfused with estrogen. ".53 Figure llc. IPSP recordings made after switching the flow ofbath solution from estrogen to normal ACSF.."".. "...""".. ".....".""...".....""""".""." 54 Figure 12. Effect ofestrogen on the early membrane resistance. """"""".55 Figure 13. Effect ofestrogen on the membrane resistance measured via late current. """,""" 56 Figure 14. Effect ofestrogen on voltage response to hyperpolarizing current .. . 57InJ ectlon . Figure 15. Effect ofestrogen on resting membrance potential. """"."""".58 Figure 16. Effect ofestrogen on action potential frequency. """"""""""" 59 Figure 17. Effect ofestrogen on the voltage response ofa BLA pyramidal neuron to an 800 pA depolarizing current. """""""""""""""""""""". 60 IX Figure 17a. Voltage Trace showing the response ofa BLA pyramidal neuron in control ACSF 60 Figure 17b. Voltage Trace showing the response ofa BLA pyramidal neuron in ACSF containing estrogen 61 Figure 18. Effect of4-0RT and the blocking effect of4-0RT on EPSP amplitude modulation 62 Figure 19. Effects of4-0RT and concurrent perfusion of4-0RT and estrogen on the early membrane resistance 63 Figure 20. Effects of4-0RT and concurrent perfusion of4-0RT and estrogen on the late membrane resistance 64 Figure 21. Effects of4-0RT and concurrent perfusion ofestrogen and 4- ORT on resting membrane potentiaL 65 Figure 22. Effects of4-0RT and concurrent perfusion ofestrogen and 4- ORT on action potential frequency 66 x Introduction Estrogen is a female hormone that appeared to protect neurons from cell death (Behl et a11995; Goodman, 1996; Green, 1996; Reibel et al., 2000; Singer, 1996, 1998) and may playa protective role against several neurological diseases such as Alzheimer's disease (Birge et ai, 1997; Henderson et ai, 1994, 1997; Kawas et al., 1997), Parkinson's disease (Bedard et al., 1977), and cerebral ischemia (Hum and Macrae, 2000; Sawada et al., 2000; Rusa et al., 1999; Fung et al., 1999). Thus, postmenopausal women have the lowest levels ofcirculating estrogen and the highest probability ofdeveloping AD. It is currently not known how estrogen protects individuals from AD, nor is the actions ofestrogen in the brain fully understood. The present study was undertaken to examine the actions ofestrogen on neuronal function and synaptic transmission in the mammalian brain. Alzheimer's disease Alzheimer's disease (AD) is a disorder ofthe central nervous system that involves neuron and synapse loss, the presence ofsenile plaques, neurofibrillary tangles, and neocortical atrophy (Reviewed by Bondi and 1 Lange, 1998). In 1907, Alois Alzheimer, a German physician and neuropathologist, first described the morphological symptoms we now know today as AD. Dr. Alzheimer studied the brain ofa patient who had died from dementia and found several distinct morphological changes in the AD brain (Reviewed by Bondi and Lange, 1998). These symptoms presently affect 50% ofall people over the age of85 and are predicted to affect 14 million people by the year 2050 (Reviewed by Birge et al., 1997). Once diagnosed, average life expectancy is 7 to 10 years, ofwhich most will be spent in a long-term care facility. In their final stages, AD patients are thoroughly debilitated by the neurological effects. AD patients experience a slow and painful decay ofall senility and eventually die while in a permanent vegetative state (Reviewed by Bondi and Lange, 1998). Neurofibrillary tangles and senile plaque formation significantly contribute to neuron loss, the subsequent decrease in synapses and neurotransmitter content, and the neocortical atrophy seen in AD patients. Unfortunately, the neuritic plaques and neurofibrillary tangles associated with AD can only be found through post-mortem staining. Ifthese morphological changes are later found in brain tissue taken from a dementia patient, there is conclusive evidence that the patient suffered from AD (Reviewed by Bondi and Lange, 1998). 2 Neurofibrillary tangles are associated with the cytoskeleton ofneurons affected by AD. Neurofibrils, also known as neurofilaments, are long strands oflinked protein subunits. Neurofibrillary tangles are formed from neurofibril subunit deformation, and this directly impacts structure and function ofthe neurofibrils (Kandel et al., , 1995). Transport proteins and structural support proteins ofneurofibrils lose their delicate organization and become a chaotic filamentous intertwining structure (Reviewed by Bondi and Lange, 1998). Neurofibrillary tangles cause death in neurons by inhibiting the efficient transport ofmaterials throughout the neuron. Neurons with neurofibrillary tangles eventually die from the lack ofessential transportation ofcellular components (Kandel et al., , 1995). Neuritic plaques are found in the areas between neurons ofsuch areas as the amygdala, hippocampus, and other cortical regions (Reviewed by Bondi and Lange, 1998). The neuritic plaques are formed by deposits of insoluble ~-amyloid protein. ~-amyloid is cleaved from amyloid precursor protein (APP), a soluble protein normally found in the brain. The soluble form ofB-amyloid is a protease released in response to neuronal injury where it plays a neuroprotective role. It is believed that an alteration in the normal metabolism ofthis protein can contribute to the development ofAD by making the cleaved ~-amyloid protein insoluble and cytotoxic (Chao et 3 al., 1998). Soluble p-amy10id has been shown to be non-cytotoxic in some cases, and insoluble p-amy10id that forms neuritic plaques has been found to be neurotoxic to hippocampal neurons in culture (Cotman, et al., 1992; Pike, et al., 1991). The toxicity ofp-amy10id may result from its actions as a protease. After cleavage oftheir substrates, proteases release free radicals as by products. Free radicals in solution can oxidize cellular components essential for a variety ofcellular processes. Free radical production is believed to cause the toxicity ofthe p-amy10id plaques (Maury, 1995). Under normal conditions, the soluble form ofB-amy10id peptide is helpful to the central nervous system, but due to a change in the position ofcleavage from its precursor protein, APP, the insoluble form ofthe peptide ironically becomes a detriment to neuronal functioning (Maury CPJ, 1995). The change in cleavage site is believed to be inherited, and this may underlie the genetic susceptibility that some families display for early onset ofAD (Chao et al., 1994). Psychological signs ofAD include unsteady development ofdementia and a gradual impairment ofmemory, judgment, and concentration (Reviewed by Bondi and Lange, 1998). Since AD is the most common cause ofdementia (Henderson., 1997), all cases ofdementia especially in the 4 elderly have a very high chance ofbeing a symptom ofAD. Dementia in AD is marked by one or more ofthese disorders: aphasia (problems with language), apraxia (problems with carrying out motor activity despite having the physical ability), agnosia (problems with object recognition despite intact sensory function), and impaired planning, abstracting, and sequencing (Reviewed by Bondi and Lange, 1998). AD patients may suffer from depression at early stages ofthe disease and they may exhibit abnormal fits ofrage as the disease progresses (Reviewed by Bondi and Lange, 1998). These fits ofrage are closely associated with other personality changes found in AD. Behavioral symptoms ofAD probably result from the neurodegeneration occurring in the limbic structures, such as the amygdala, as well as in higher cortical areas (Reviewed by Bondi and Lange, 1998). Treatment ofAlzheimer's Disease Currently, a suggested means oftherapy and prophylaxis for AD patients is the administration ofvitamins A, C, and E (Reviewed by Bondi and Lange, 1998). These vitamins are good antioxidants, which act to subdue the cytotoxicity of~-amyloid. Vitamin E (a-tocopherol) has shown 5 to playa protective role against the neurotoxic effects of ~-amy10id in animal models (Jaffe et al., 1994), , and it also shows the potential of delaying AD due to its strong antioxidant activities (Birge et al., 1997). The antioxidants absorb the free radicals generated by ~-amy10id, and other sources offree radicals such as cyclo-oxygenase, a chemical released upon an inflammatory response. Some functions ofcyclo-oxygenase include involvement in the production ofprostaglandins and mediation of excitotoxic cell death via the glutamatergic N-methy1-D-aspartate receptor. Anti-inflammatory drugs such as indomethicin can reduce the risk of developing AD by 50% (Birge, et al., 1997). Several currently prescribed AD medications stop the actions of enzymes at the axon terminal from cleaving used neurotransmitter. Two of the most commonly prescribed medications for AD at this time are donezapi1 (Aricept) (Birge et al., 1997) and tacrin (Cognex) (Henderson et al., 1997). These medications have been reported to slow the onset ofAD symptoms by inactivating cho1inesterases (Birge et al., 1997). These enzymes are responsible for breaking down acetylcholine at cholinergic synapses. The patients receiving donezapi1 experience an increase in the amount of acetylcholine at synapses (Birge et al., 1997) between intact cholinergic 6 basal forebrain neurons and efferent neurons such as those located in the basolateral amygdala, hippocampus, and higher cortical areas. Estrogen has also been suggested as a treatment for AD (Birge et ai., (1997). Neuroprotective effects ofestrogen have been implicated in studies ofcytotoxicity (Behl et ai, 1995: Goodman. 1996; Green et ai., 1996; Singer et ai., 1996, 1998), and it has been shown that estrogen increases blood flow to the brain during clinical trials with AD patients (Ohkura et ai., 1994). In addition, estrogen replacement therapy has been suggested, through multiple epidemiological studies, to possibly delay AD onset and slow cognitive decline in patients experiencing AD symptoms (Brenner et ai., 1994). Heredity ofAlzheimer's Disease Genetic research has shown that heredity plays a large role in the likelihood ofindividuals to develop AD. The chance ofdeveloping AD increases four times ifa first-degree relative has the disease, and it has been found that 80% ofall AD occurrence is attributed to heredity (Birge et ai., 1997). The additional 20% may be caused by environmental factors such as head injuries and chemical insults that may have occurred at a time as far back as childhood (Birge et ai., 1997). The inherited trait that represents 7 individual susceptibility to AD development is believed to be the ApoE epsilon-4 allele (Holtzman et al., 1995). ApoE protein is normally released in response to the neurodegeneration and other neuritic insults occurring in the brain (Mahley, 1988; Boyles et al., 1989; Boyles et al., 1990; Poirier et al., 1991)). AD patients have been tested for the mutated ApoE protein. It has been found that people who have experienced head trauma with the "bad" ApoE epsilon-4 allele have been found to develop AD at a greater frequency than those with the wild-type ApoE gene (Mayeux et al., 1995) . A malfunctioning ApoE protein is not able to help the neurons recover from injury. Normal functioning ApoE protein helps the injured neurons by making cholesterol available for cell membrane repair. Correspondingly, neurons located in areas ofthe brain affected by AD contain a low amount ofcholesterol, and the delivery ofcholesterol to these regions is markedly impaired (Mason et ai, 1992). Estrogen Estrogens represent a class ofseveral closely related steroid hormones, ofwhich 17-~-estradiol (estrogen) is the major form synthesized 8 by the ovary (Johnson and Everitt, 1995). Synthetic pathways for estrogen are numerous (Fig. 1). Aromatase is an important enzyme in this pathway because it helps to control local estrogen blood levels (Johnson and Everitt, 1995). This enzyme has a wide distribution throughout different tissues in the body; including brain tissue (Balthazart and Ball, 1998). The mammalian limbic and hypothalamic regions contain high levels of aromatase (Balthazart and Ball, 1998). Activity ofthis enzyme is controlled in the ovarian cells by local blood levels offollicle stimulating hormone (Johnson and Everitt, 1995). Follicle stimulating hormone and luteinizing hormone are secreted from the anterior lobe ofthe pituitary gland, and control the blood levels ofestrogen in a negative feedback loop with blood levels ofestrogen (Johnson and Everitt, 1995). Aromatase is responsible for the aromatization oftestosterone into estrogen and the aromatization of androstenedione into estrone (Balthazart and Ball, 1998). Estrone is an estrogen easily converted to l7p-estradiol (Johnson and Everitt, 1995). Thus, the presence ofaromatase in other tissues gives those tissues the chance to capitalize on an extra dose ofestrogen ifblood levels of testosterone are at a concentration to permit aromatization. Luteinizing hormone participates in the regulation ofestrogen synthesis earlier in the pathway. The levels ofthis hormone modulate the rate ofpregnenolone, 9 formation from cholesterol (Johnson and Everitt, 1995). Pregnenolone is a steroid precursor for testosterone/estrogen synthesis (Johnson and Everitt, 1995). 10 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 I (}Hc;b/OH tfl)/ctj 16ahydroxytesterone OIl -o~~ 16a hydroxy androstenediol ? ISOm~'i1!14! oo:n?? . 1111I'" . IEstramol I ,,~~ ')(1 t...l1uO;rt." ort? 16a hydroxyandrostenedione 5a dihydrotesterone .. ~ ~ HoJ.hl Alomata!l4!t u ~~ ~ ?dPo 0 i I \111?14 $ (=001.. 110 HO" >.. i - Progesterone 17a hydroprogesterone 1 1 C~J (!,') H(-OH 11(-011 ~ # I I 20a dihydroprogesterone The Genomic Pathway ofEstrogen Action Estrogen has traditionally been described as acting on tissues through a slowly developing and long-lasting mechanism commonly referred to as the genomic pathway. In this pathway, estrogen influences the cell by initially binding to an estrogen receptor (ER) , which is found at highest concentration in the cell nucleus (Li et at., 1997). After becoming activated, the following sequence ofevents has been reported to occur (Reviewed by Tsai and O'Malley, 1994). The estrogen-receptor complex can interact with transcription factors. These transcription factors regulate specific sequences ofDNA. In the absence ofestrogen, the ER exists bound to heat shock proteins and remains inactive. After estrogen binding, the receptor undergoes a conformational change at its ligand-binding domain, and the receptor becomes activated at protein activating functions. This leads to dissociation ofthe receptor from the heat shock protein followed by ER dimerization, the coupling oftwo ERs. To activate the ER, the receptor must be in the presence ofestrogen or a receptor agonist. Removal ofthe heat shock proteins through other means is not sufficient for activation ofthe ER. After the ER dimerizes, the ER is now able to bind to a specific DNA complex and interact with transcription factors to activate or inactivate the target gene. Gene activation leads to mRNA production and increased 12 synthesis ofa specific protein that is then responsible for production ofthe estrogen-induced cellular response. The period between activation ofER mediated transcription and post-translational modification ofthe final protein product may involve hours or days . The Estrogen Receptor (ER) There are two well-described estrogen receptors, ERa and ERB, although some evidence points to the possibility ofa third receptor type (Kuiper et al., 1997). ERa and ERB belong to a superfamily of steroid/thyroid receptors. Within this nuclear receptor superfamily, estrogen receptors belong to the largest subgroup that also includes glucocorticoid receptors, androgen receptors, progesterone receptors, and mineralocorticoid receptors. There is a high degree ofconservation amongst these receptors in their hormone-binding and DNA-binding domains (Kuiper et al., 1997; Tsai and O'Malley, 1994; Wrenn, 1993). In rat and human neurons, ER and other androgen receptor proteins have been detected in neurons (Couse et al., 1997) with distributions from nuclei to far reaching cytoplasmic extensions such as dendrites and axon terminals (Greco et al., 1998; Puy et al., 1995). In the rat brain, neurons 13 containing ERB and ERa are distributed together in several cortical amygdaloid nuclei (Puy et al., 1995) and some areas ofthe hypothalamus (Shughrue et al., 1998; Weiland 1997). ERB, without the presence ofERa, is broadly expressed throughout the rat brain. Both isoforms ofER may have different actions in cells because oftheir different DNA binding domains (Shughrue, et al., 1998). Complex cellular responses can arise because both ERa and ERB can dimerize with their homologue or heterodimerize with the other type ofER (Katzenellenbogen et al., 1996). When ERs bind to DNA, both DNA recognition sites ofthe dimerized ERs recognize their own specific sequence. When heterodimerization occurs it is possible to have differences in function due to the differentiation in binding characteristics ofERa and ERB (Shughrue, et al., 1998). The ER protein is large protein with a number offunctionally important motifs along its chain ofamino acids. cDNA evidence indicates that ERB consists of485 amino acids with a calculated molecular weight of 54,200 Daltons. Isomer homology at the ligand-binding domain ofERs is about 90%. Homology between DNA binding motifs is 50% (Kuiper, et al., 1997). The differences between ERa and ERp have developed to allow different receptors to activate different areas ofthe genome while being activated by a common hormone. 14 Antagonism ofthe ER Estrogen replacement therapy increases the incidence ofdifferent types ofcancer in female reproductive tissues. Extensive research with uterus and mammary tissue led to the discovery ofantagonists to inhibit some ofthese side effects ofestrogens. Drugs such as Tamoxifen, ICI, and Raloxifene competitively inhibit the estrogen receptor, and thus decrease the chances ofcancer developing in some peripheral tissues (Gradishar and Jordan, 1997; Jordan, 1998). However, both Tamoxifen and Raloxifene are known to also have agonistic actions on the ER (Berry and Metzger. 1993; Ali et al., 1993; Metzger et al., 1995). In addition, the agonistic actions of these drugs differ from each other. Cellular responses to estrogen are very complex, and have yet to be completely defined. Understanding how the separate regions ofthe ER function while initiating cellular changes is integral to understanding the intricacies ofER antagonism/agonism duality. Antagonism for the ER is an important part ofmodem medicine. To have an effective ER antagonist is to have the best ofboth worlds; drugs that can block the cancer associated with estrogen without reducing the beneficial actions ofestrogen. It is not unusual for ER antagonists to have 15 more than one action in patients. The selective estrogen receptor modulator (SERM) tamoxifen is an ER antagonist, which is regularly prescribed for those considered to be at high risk for developing breast cancer. Chances of developing breast cancer drop by 45% after tamoxifen treatment (Smigel, 1998). At the same time, bone density increases so that fewer fractures occur, which is an important benefit ofestrogen replacement therapy. However, there is an unfortunate increase in endometrial cancer incidence (Smigel, 1998). This phenomenon ofinactivation/activation or antagonism/agonism is further exemplified by postmenopausal women experiencing increases in bone density when receiving the SERM raloxifene for treatment ofosteoporosis (Black et al., 1983). Understanding how SERMs work in different tissues and considering their actions on the individual estrogen receptor types (ERa and ERB) will help us to find clues that will eventually lead to the finding ofa SERM with the ability to mimic the positive estrogenic influences but block the carcinogenic responses. Defining the mechanism by which SERMs selectively activate and inactivate the ER is key in understanding how these chemicals can be both an asset and a liability. Activating functions (AF-l and AF-2) are regions of the ER, which are activated following a specific conformational change by the ER (Jensen EV et al., 1973). It is this conformational change in the ER 16 that is altered or inhibited by tamoxifen, raloxifene, and similar antagonists (Jordan, 1998). AF-l is located in the N-terminus ofthe ER (Kumar et al., 1987). AF-2 is located in the ligand-binding domain (Kumar et al., 1987). The conformational change needed to activate AF-2 is a folding ofhelix 12 ofthe ER to uncover the AF-2 domain (Shiau et al., 1998). This folding of helix 12 on the ER is inhibited by ER antagonists, and thus the AF-2 domain remains inactive (Shiau et al., 1998). This does not mean that the other ER activating function is inactive. Helix 12 is sterically hindered by an alkyl amino-ethoxy side chain ofthe antagonist molecule that is otherwise structurally similar to estrogen (Jordan, 1984). Although the AF-2 domain ofthe ER is inhibited by ER antagonists, the AF-1 region may remain active through mitogen-activated protein kinase pathways (MAPK) (Kato et al., 1995). When the AF-1 region is activated by SERMs, it initiates some ofthe same cellular changes as estrogen. Itis ironic that treatment with antiestrogens invokes some estrogenic responses. Non-Genomic Mechanism ofEstrogen Action Until recently, estrogen was thought to exert its action only via the genomic (nucleic acid! protein synthesis) pathway, a process that requires 17 hours or longer to produce a cellular response. However, rapid effects of estrogens and other steroid hormones were found in frog and fish oocytes in the 1980's. (Zakon., 1998) Since these initial reports, additional studies have shown similar rapid cellular responses to estrogen and androgens in several tissues, including the brain (Balthazart et al., 1998; Gorzynska and Handelsman., 1995; Woolley., 1999). These recent findings ofestrogenic and androgenic actions taking place in minutes or even seconds indicate that the traditional genomic pathway is not the only mechanism by which steroid hormones may exert their actions. The problem we now face is discovering a new pathway by which estrogen can function in a much shorter time frame. An example ofthe rapid action ofsteroidal hormones is seen in Sertoli cells ofthe rat testis (Gorzynska and Handelsman., 1995). Testosterone treatment causes a rapid (20-40 second) rise in cytosolic calcium levels. An identical rapid cellular response was also seen during the application of testosterone that had been conjugated to a large protein. (Gorzynska and Handelsman., 1995). Since the known steroid receptors are found in the cytosol and the nucleus, their activation depends upon the steroid penetrating the cell membrane and reaching their ligand-binding domain. With a protein attached to the steroid hormone, diffusion across the cell membrane is 18 impaired. Actions such as those described in the Sertoli cells ofmale rats (Gorzynska and Handelsman., 1995) strongly suggest the presence ofa cell surface steroid receptor. Since androgen receptors are very closely related to ER in structure and function, the existence ofandrogen membrane receptors in rat Sertoli cell membranes suggests that cell surface membrane receptors for estrogen may also exist. Another possibility for the rapid action ofestrogen is that the known receptors (ERa and/or ER~) are working in a fashion other than the genomic pathway. Signal transduction pathways are a common mechanism for receptors and some evidence suggests that estrogen receptors may evoke their rapid actions via signal transduction mechanisms. Pyramidal neurons in the hippocampus are remarkably similar to those found in the BLA. In CA1 neurons ofthe hippocampus, estrogen activates the cAMP pathway to induce AMPA/kainate currents, which are normally activated by glutamate (Gu and Moss., 1996); similarly, in neuroblastoma cells estrogen initiates a mitogen-activated protein kinase (MAPK) cascade ofevents (Watters et al., 1997). Growth factors such as epidermal growth factor and insulin-like growth factor also act through the MAPK pathway. These factors enhance the genomic response caused by estrogen, suggesting the possibility that the pathways ofaction for these chemicals may be intersecting. 19 In the MAPK pathway, there is a membrane receptor-associated tyrosine kinase initially activated by a growth factor, and the resulting cascade includes activation ofthe proteins ras, raf, and MAPK respectively (Hill CS and Treisman, 1995; Pelech SL and Sanghera, 1992: Sanghera, 1992). Introduction ofthe activated forms ofany ofthese proteins into the cytosol ofa cell had the effect ofincreasing ER activity at the AF-1 domain much like introduction ofthe growth factors (Kato et al., 1995). Further observation showed that the Ser-118 residue ofthe ER was phosphorylated following activation ofthe MAPK cascade (Kato et aI, 1995). Phosphorylation ofthis serine residue is important for the activation ofthe AF-l domain ofthe ER. Introduction of4-hydroxy-tamoxifen, an active form oftamoxifen, into the cytosol initiates phosphorylation identical to that ofestrogen, whereas the stronger antagonist ICI 164,384 does not initiate a response (Kato et al., 1990). Not only is this a possible pathway ofSERM agonism, but this information also suggests that a possible method of blocking estrogen action is through the blocking ofphosphorylation ofthe estrogen receptor. The Basal Lateral Amygdala (BLA) 20 The basolateral nucleus ofthe amygdala (BLA) is one ofseveral nuclei that make up the amygdala, an almond-shaped region located deep within the temporal lobe ofthe cortex (Martin, , 1996). As part ofthe limbic system, the amygdala plays important roles in emotion and memory formation (Martin, , 1996). The BLA consists primarily ofpyramidal type neurons that are very similar in structural and functional characteristics to neurons ofthe hippocampus (Washburn and Moises, 1992b), another important component ofthe limbic system (Martin, , 1996). The amygdala and the hippocampus receive afferent innervation from the basal forebrain (Emson et aI., 1979; Woolfand Butcher, 1982; Carlsen et al,. 1985). The BLA is the amygdaloid region that has been shown to receive the highest density ofcholinergic inputs in the region (Ben-Ari et al., 1977; Hellendall et al., 1986). Functionally, this region is thought to be responsible for giving emotional significance to memories (Martin, , 1996). Cholinergic inputs from the basal forebrain begin to decrease in number in AD. The cause ofthe decreased amount ofcholinergic inputs in AD is the death ofafferent basal forebrain cholinergic neurons (Bartus et al., 1982). The BLA is a region intimately related to the etiology ofAD, in fact it is one ofthe first regions ofthe brain to show the evidence of neurodegeneration upon the onset ofAD. The many psychological 21 dysfunctions associated with AD such as memory loss, emotional instability, and depression may originate from this region's incapacity to function properly following AD-inflicted neuronal cell death (Bartus et al., 1982). A major afferent pathway to the BLA is the external capsule (EC). This pathway carries both cholinergic and glutamatergic inputs to BLA neurons (Washburn and Moises, 1992). Briefelectrical stimulation ofthe EC evokes in BLA neurons a rapid excitatory postsynaptic potential (EPSP) response, which is often followed by a delayed inhibitory postsynaptic potential (IPSP) response. Neurons initiate EPSPs from direct innervation via EC. However, in the case ofan IPSP, neurons receive their signal via an interneuron. Interneurons receive excitation from a presynaptic source such as the EC, and generate an IPSP in postsynaptic cells (Rainnie et al., 1991a, b). A long-lasting depolarization ofBLA neurons is mediated by the synaptic release ofacetylcholine, but this response is only seen following high frequency EC stimulations (Washburn and Moises., 1992c), which are not used in the present study. Clinical evidence suggests that estrogen may playa protective role against the onset ofAD (Birge et ai, 1997; Henderson et ai, 1994, 1997; Kawas et al., 1997). Estrogen has also been reported to have effects on cognition (Birge et al., 1997; Ripich et al., 1995) and long-term potentiation 22 (Cordoba and Carrer, 1997). Since the amygdala is an early and severely affected target ofthe neurodegeneration associated with AD, it was of interest to investigate the actions ofestrogen on neurons ofthis region. This project was thus undertaken to test whether estrogen altered the functional properties ofBLA neurons or their synaptic inputs. 23 / External Capsule ~-- Stimulator Presynaptic Stimulating Electrode---- ( BLA Inhibitory Interneuron Amplifier, computer, and oscilloscope Ground Electrode BLA Nerve cell Materials and Methods Long-Evans rats ofrandom sex and aged 3 weeks to 1 year were used in this study. Rats were housed on 12 hour light/dark schedule and received food and water ad libitum. All procedures involving live animals were approved by the Animal Care and Use Committee, Youngstown State University. Procedures were modified from Washburn and Moises (1992b). Animals were sacrificed by decapitation, their brains removed promptly, and placed in oxygenated and ice-cold artificial cerebrospinal fluid (ACSF). The ACSF was made in 1.5 liter batches daily including the following constituents with concentrations in millimolars 124 NaCl, 3.5 KC1, 1.5 MgS04, 1.0 anhydrous monobasic NaH2P04, 26.2 NaHC03, 11.0 glucose, and 3.0 CaCho Three to five slices ofventral forebrain containing the amygdala were cut on the horizontal plane at a thickness of400 Jlm using a Vibroslicer (Campden Instruments). The slices were held at room temperature (23?C) in ACSF bubbled continuously with 95% O2/ 5% CO2 for one hour before any recordings were attempted. This assured time for the cells to recover from the vibrotome. Individual slices were placed the recording chamber as needed. The slice was held between two layers of nylon mesh submerged in continuously flowing ACSF at room temperature. 25 Intracellular recordings were obtained from neurons ofthe BLA using glass microelectrodes filled with 2.7M KCII OAM K acetate (pH 7.0) and having resistances of70-120 MOhms. Recordings were amplified with the Axoclamp 2B in Bridge mode (Axon Instruments) and displayed on a Tektronix 2213A oscilloscope and Cole Parmer chart recorder. Signals were also fed to a computer interface (INDEC, Sunnyvale California) that digitized the analog signal for storage and analysis by a microcomputer based program (pCLAMP, Axon Instruments). Only cells with membrane potentials greater that -55 mV and over shooting action potentials were included in this study. Under our recording conditions, the vast majority ofBLA cells were not spontaneously active but cells could be made to fire by passage ofdirect depolarizing circuit through the electrode. Neuronal input resistance was determined by passing an incremental series ofcurrent pulses (0.2 nA increments; range, -1.0 nA to +0.4 nA; 450 msec duration) through the recording electrode and measuring the resultant voltage deflections. EPSPs were elicited in BLA neurons by delivering voltage pulses of 0.1 msec duration originating from an A31 0 Accupulser (World Precision Instruments), isolated via an A360 Stimulus Isolator (World Precision Instruments), and introduced through a bipolar stimulating electrode placed 26 on the surface ofthe slice over the external capsule (EC). While the EC could be easily identified visually, we relied on anatomical landmarks in the slice and diagrams ofstereotaxically defined sections (Paxinos and Watson, Academic Press, 1998) for guiding placement ofthe recording electrode in the BLA. Typically, the stimulus intensity delivered was that which evoked an EPSP whose amplitude was just below threshold for generating an action potential when recordings were made at the resting membrane potential. Membrane potential was manually adjusted by intracellular injection of direct current through the recording electrode, and was held at -65 to -75 mV when characterizing EPSPs elicited by EC stimulation. Estrogen (17- ~-estradiol) (Research Biochemical International) and/or 4-0H-tamoxifen (Calbiochem) were applied to the slice by switching the bath superfusate from normal ACSF to ACSF containing known concentrations ofthe drugs. A 2 IlM Estrogen concentration in ACSF was prepared by the addition of 1.2 ml ofdaily-made 100 IlM stock solution to 60 ml saline. A 60 nM 4-0H-tamoxifen solution was prepared by the addition of12 ilL ofO.061lM stock solution stored at O?C to 60 ml saline. All stock solutions were prepared with 95% ETOH as a solvent. Tests were performed on neurons that passed criteria listed above for membrane health. Tests were conducted in a series, which included current 27 stimulation, single-pulse presynaptic stimulation, and train presynaptic stimulations. Voltage recordings were taken for all tests in the series, and some recordings were used in determining resting membrane potential. To decipher the early and late membrane resistances, the injected current and voltage responses were used in accordance with Ohm's Law. EPSP amplitudes were measured from resting membrane potential to the maximum point on the voltage traces resultant ofsingle-pulse stimulation. The number ofaction potentials fired was counted during the 450 ms current stimulation at 800 pA ofdepolarizing current. Accommodation was analyzed by comparing experimental recordings to control. The results ofthe experiment were analyzed for statistical significance using a two-tailed t-test. During the course ofthe experiments involving estrogen, the bath superfusate was changed from control saline to estrogen and back to control saline for washout. During the course ofexperiments involving 4 OH tamoxifen, the bath superfusate was changed in the following manner: control, 4-0H-tamoxifen, control for washout, saline containing estrogen and 4-0HT (to test blocking), control saline for washout, and saline containing estrogen. The series oftests were conducted for 20 minutes after switching the bath to the next solution in sequence. New slices were placed in the recording chamber at the conclusion ofan experiment. 28 Results The experiments presented here were designed to investigate the actions ofestrogen on synaptic transmission in the BLA. Perfusion ofBLA neurons with ACSF containing estrogen (2 f.lM) resulted in a significant decrease in excitatory transmission and EPSP amplitude (Fig. 3). The mean EPSP amplitude in control ACSF was 9.9 ? 2.8 mV (? SD; n=7). Approximately 20 minutes after the bathing saline was changed to ACSF containing estrogen, mean EPSP amplitude was reduced to 0.8 ? 1.2 mV (n=7, p <0.001 from control). Switching the saline flow back to control ACSF was followed by a complete recovery ofevoked EPSP amplitude, which returned to a mean amplitude of9.6 ? 3.4 mV (n=7), a value not significantly different from the control amplitude. When observing EPSP recordings (Fig. 4b), the change in EPSP amplitude upon addition ofestrogen is seen as a gradual decrease ofEPSP amplitude within 20 to 30 minutes. Frequently (4 out of7 cells), estrogen completely eliminated an evoked EPSP. Upon wash, the evoked EPSP amplitude recovered and the cell once again showed normal synaptic responses within 30 minutes. Spontaneous synaptic activity was also reduced in the presence ofestrogen (Fig. 5a-c). The level ofspontaneous 29 activity in control cells (Fig. 5a) is higher than that observed for cells in the presence ofestrogen (Fig. 5B). Typically, cells with lower EPSP amplitudes had correspondingly lower levels ofspontaneous activity. Effects ofethanol, the vehicle used to dissolve estrogen and tamoxifen, on EPSP amplitude were studied as a control against the possibility ofethanol playing a role in the effects ofestrogen on EPSP amplitude (Fig. 8). Mean EPSP amplitude in the presence ofACSF containing 2% ethanol was 9.0 ? 1.4 mY compared to mean control EPSP amplitude of9.9 ? 2.9 mY (n=2). These findings indicate that alcohol alone has no effect on EPSP amplitude. Effects ofestrogen on synaptic transmission were also evident during multiple (train) stimulations ofthe presynaptic pathway (n=9). When the recordings oftrain stimulation are superimposed, the effects ofestrogen in comparison to control ACSF are visible as a lower amplitude response to the stimulation (Fig. 6a). The reversibility ofthe response to estrogen is seen as a return ofthe control train response to normal (Fig. 6b). Same cell traces of train stimulation responses in the presence ofcontrol ACSF (Fig. 7a), estrogen (Fig. 7b), and wash (Fig. 7c) confirm the effect ofestrogen on responses to train stimulation. 30 Preliminary data suggested that estrogen did not alter IPSP amplitude. When comparing IPSP amplitudes in control ACSF and ACSF plus estrogen (Fig. lOA), there appeared to be no noticeable difference. Individual traces in figures 11 a-c further emphasize the similarity ofIPSPs in the presence ofnormal ACSF, estrogen, and wash. Estrogen appeared to have no effect on early membrane resistance (Fig. 12), late membrane resistance (Fig. 13), resting membrane potential (Fig. 15), number ofaction potentials (Fig. 16), or accommodation response (Fig. 17). Membrane resistance was calculated from the size ofthe voltage deflection produced during the intracellular injection ofa 450 ms, 400 pA hyperpolarizing current pulse. BLA pyramidal neurons normally respond to a hyperpolarizing current with an initial peak negative voltage response, followed by depolarizing (upward) voltage sag (Fig. 12) This is consistent with studies ofneurons characterized in this region (Washburn and Moises, 1992b) The depolarizing sag is due to the hyperpolarization-induced activation ofoutward current (Womble and Moises, 1992). The input resistance ofBLA neurons was thus determined at 2 time points: at the initial peak ofthe voltage response (early resistance) and just prior to the end ofthe current pulse (late resistance). Estrogen had no significant effect on 31 either the early or late membrane resistances. Mean early membrane resistance for control neurons in the presence ofcontrol estrogen and after estrogen washout were 61.5 ? 15.6 MO, 59.0 ? 13.0 MO, and 70.2 ? 9.9 MO respectively. These values are consistent with previous reports (Washburn and Moises, 1992b; Womble and Moises, 1992; Gean and Shinnick-Gallagher, 1988, 1989) Mean late membrane resistances for neurons in the presence ofcontrol ACSF, estrogen, and wash were 53.5 ? 13.6 MO, 52.0 ? 14.4 MO, and 63.4 ? 11.7 MO respectively. Estrogen appeared to have no effect on the resting membrane potential ofBLA neurons. Mean values ofresting membrane potentials recorded in control ACSF, estrogen, and wash were not significantly different. Mean resting membrane potentials recorded in the presence ofACSF, estrogen, and wash are -62.6 ? 6.3 mY, -64.8 ? 7.5 mY, and -62.5 ? 9.3 mY respectively. Estrogen also appeared to have no effect on action potential frequency in BLA neurons. Action potentials are easily distinguished by a sharp depolarization followed by a sharp hyperpolarization. Action potential frequency was measured by the number ofaction potentials generated during 450 ms pulse of800 pA depolarizing current. In control ACSF, the mean action potential number was 5.9 ?2.5 APs. Cells in the presence ofestrogen 32 and wash had mean AP numbers of4.6 ? 3.0 APs and 7.8 ? 2.2 APs respectively. In addition, the recordings were examined for any change in accommodation response (fig. 17). Accommodation response is seen as a relative change in the frequency ofaction potential firing over time. Upon examination, no significant differences were found in the accommodation response ofneurons studied under saline flow containing control ACSF and estrogen. Additionally, no changes were found when the bath was switched from ACSF containing estrogen back to normal ACSF. To test the specificity ofthe estrogen response, an estrogen receptor antagonist was used to block estrogen receptors. The chosen antagonist, 4 OHT was used at 60 nM concentration. This concentration was used because 4-0HT's binding affinity for ERa is over 300 times that ofestrogen (Kuiper et al., 1997) The effects ofthis drug on BLA neurons (n=2) were examined to study the drug's blocking and direct effects on the cell. These preliminary results may point to possible roles estrogen has in the brain. 4-0HT has the effect ofsignificantly increasing the EPSP amplitude (p < 0.05)(Fig. 16). Mean EPSP amplitude for recordings made in the presence of4-0HT are 15 ? 0 mY. This is significantly greater than the mean ofEPSP amplitudes recorded in control ACSF (9.9 ? 2.9 mY, p < 0.05). 4-0HT prevented the inhibitory effect ofestrogen on EPSP 33 amplitude. Concurrent 4-0HT and estrogen perfusion gave no significant change in the EPSP amplitude (mean= 9.8 ? 4.6 mY). 4-0HT alone or concurrent perfusion of4-0HT and estrogen appeared to have no effect on early membrane resistance (Fig. 17), late membrane resistance (Fig. 18), resting membrane potential (Fig. 19), frequency ofaction potential number (Fig. 20), or the accommodation response (not shown). 4-0HT and concurrent perfusion of4-0HT and estrogen had no significant effect on the early or late membrane resistances. In control ACSF, the mean value ofearly membrane resistances was 63.8 ? 14.8 MO. Cells in the presence of4-0HT and concurrent perfusion of4-0HT and estrogen had mean early membrane resistances of54.0 ? 8.5 MO and 62.5 ? 16.3 MO respectively. The mean value oflate membrane resistances in control ACSF was 62.6 ? 6.3 MO Cells in the presence of4-0HT and concurrent perfusion of4-0HT and estrogen had mean late membrane resistances of62.5 ? 7.2 MO, and 72.5 ? 3.5 MO respectively. 4-0HT and concurrent perfusion of4-0HT and estrogen also appear to have no effect on the resting membrane potential ofBLA neurons. In control ACSF, the mean value ofresting membrane potentials was -62.6 ? 6.3 mY. Cells in the presence of4-0HT and concurrent perfusion of4-0HT 34 and estrogen had mean resting membrane potentials of-62.5 ? 3.5 mY and 72.5 ? 3.5 mY respectively. These values were not significantly different. 4-0HT and concurrent perfusion of4-0HT and estrogen appeared to have no effect on action potential number. In control ACSF, the mean number ofaction potentials was 5.9 ? 2.8 APs. Recordings in the presence of4-0HT and concurrent perfusion of4-0HT and estrogen had mean action potential numbers of5.0 ? 2.8 APs, and 3.0 ? 1.5 APs respectively. In addition to action potential frequency, the recordings were examined for any change in accommodation response. Upon examination, no noticeable differences were found in accommodation response ofneurons studied in control ACSF, ACSF plus 4-0HT, or concurrent perfusion of4-0HT and estrogen (Fig. 17a, b). 35 j j j j j j j j j j j j j j j j j j j j j j j j j j j j l Effect of Estrogen on EPSP Amplitude 14 T -----_..~ ~ IllllJ Control J T ACSF 12 -j rn Estrogen 9.9mV 10 I 9.6mV III Wash :;-.s 8 CD "0:e 0..E ? 6a.. (f)a.. w 4 2 o+--------- I j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j >E o T""" (/)E oo T""" N ....lIo.o o 3en ....lIo.o 3< U1o 3en (]'Io 3en (J'l ~ 0"o - 3< U) Eo L{) >E -. 0U T""" LO -<0 >E o-r-- enE oo L() j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j * CJ'1o o 3 CJ) ....Ji,.o 3< 0> C'"- I\.) CJ1o 3en -lo.o 3< * NCJ1 o 3en ...lIo.o 3< * N01 o 3en ~o 3< j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j Effect of Ethanol Vehicle on EPSP Amplitude 14 -"----------------- 12 I ---------tl------------------- 11IIII Control ACSF 9.9mV 10 +1-------------, :;-.s all- _ CD"0 :::J:!: "i5..E ~ 6 1--------(f) a..w 4 +1------------j 2 +I---------~ o+1--------- 9.8mV rn Ethanol and ACSF I j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j ....ll.o o 3en ....ll.o 3< CD Q)- 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 ...Jo.o o 3(J) CD0'" '--" ~o 3< j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j ~o 3< ~o 0"--- ~o 3< ~o 3< -J.o 3< Effect of Estrogen on Early Membrane Resistance 90 '",----. 80 70 60 Ii) E.r:. o 50 ~ Q) (,)c: .l9 401Il "iii Q)0:: 30 -I . 20 10 o-'-,---------- 61.5 MOhms 59.0 MOhms 70.2 MOhms IIIllJ Control ACSF rn Estrogen []]I Wash 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 I 80TI~~~~~ Effect of Estrogen on Late Membrane Resistance IlJIl) Control ACSF 70 60 50 40 30 20 10 O-'--I~~~~~ 53.5 MOhms l 52.6 MOhms 63.4 MOhms mEstrogen m1Wash j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j - 10 1 --- E- Q) "0:E c.. 8 1 ~ 0 C/) 0- W 6 I 4+1------- 2 +1-------- o+1-------- 9.8mV rn4-0HT and Estrogen 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 1 I Effect of 4-0HT and Estrogen Blocking Effect of 4-0HT on Early Membrane Resistance 90 1 IIlIIIJ Control ACSF 80 70 Ii) 60E .co ~ ~ 50c ~ "iii CDc:: 40 CDc e!.0 ~ 30::2: 20 10 o.."-1 _ 1 63.8 MOhms T 54.0 MOhms I 62.5 MOhms m4-0HT rn4-0HT and Estrogen Effect of 4-0HT and Estrogen Blocking Effect of 4-0HT on Late Membrane Resistance 80,,~~~~~~ 70 1 "T" I IllllJ Control ACSF rnJ4-0HT 60 1-?---?__?-55.3 MOhms lilE .s:::.o 50 6 ~c: ~.~ 40 130::: Q)c: ~~ 30 rl~-~~~~- Q) ~ 20 +1~~~~~~~- 10+1~~~~~~ O..LI~~ ~ 55.0 MOhms 56.3 MOhms rn4-0HT and Estrogen Effect of4-0HT and Estrogen Blocking Effect of 4-0HT on Resting Membrane Potentialo -10 +1------- -20 +I------~ >5 -30 ;m c::2 ~ -40 1 Q)c:: e!.0 E~ -50 I I -60 +I------~ -62.5 mV -70 I - -72.5 mV -80 -'-1 _ IIIllI Control ACSF rn4-0HT rnI4-0HT and Estrogen j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j j Effect of 4-0HT and Estrogen Blocking Effect of 4-0HT on Action Potential Frequency 10 I ------- ~ Ililll Control ACSF 9 -t-I----------------------------------------------j m4-0HT 8 I -----+1-----------------------------1 rn4-0HT and Estrogen 5.0 5.9 - 1111111111111111111111111111111111111111111111111111111111111111111111111111111111111111111111111111111111111 3.00L- Q) 3 .0E ::JZ 2 1 0 I Q) IJ) ~ 71 IJ) Eo o '24 hours), and were not found in the BLA (Canonar et al., 1993). Therefore these results 72 (Canonaro et ai., 1993) should have few implication on the synaptic alteration found in BLA neurons. Cholinergic responses are elicited in the postsynaptic cell by the release ofacetylcholine from presynaptic cholinergic neurons. Their axons run within the EC and are activated following EC stimulation (Washburn ad Moises, 1992a, b). Cholinergic receptors on BLA neurons are activated by acetylcholine release following high frequency stimulation ofthe presynaptic external capsule pathway (Washburn and Moises., 1992b, c; Rainnie et ai., 1991a). Cholinergic input to cells in the BLA arises, for the most part, from regions outside ofthe BLA (Emson et ai., 1979; Woolfand Butcher., 1982; Carlsen et ai., 1985), although there is evidence showing small populations ofcholinergic neurons in the BLA (Carlsen and Heimer, 1986). The cholinergic axons found in the EC have been traced back to cholinergic neurons residing across a broad region that encompasses the diagonal band ofBroca to the ventral palladium (Emson et ai, 1979; Woolf and Butcher, 1982; Carlsen et ai., 1985). Acetylcholine can elicit excitatory responses in BLA neurons via the muscarinic type ofcholinergic receptor (Washburn and Moises., 1992a). Cholinergic responses consist of a long, sustained depolarization ofthe postsynaptic BLA neuron (Washburn and Moises, 1992a). The effect ofestrogen on the acetylcholine pathway was 73 not a focus ofthis study. However, pursuit ofinformation regarding how estrogen interacts with the acetylcholine pathway remains an area ofgreat interest. Acetylcholine and glutamate pathways are the two known and described excitatory pathways to the BLA. In the present study, EPSP amplitude and spontaneous activity decreased upon addition ofestrogen (Figs. 3,4,5). Glutamate is the neurotransmitter associated with EPSP elicitation. Thus, the cause ofsynaptic activity alteration in BLA neurons is likely associated with the glutamate system. The receptors involved in the estrogenic alterations ofsynaptic activity were examined with the use ofthe ER antagonist 4-hydroxy tamoxifen (4-0HT). Non-steroidal ER antagonists such as 4-0HT are structurally similar to estrogen with the exception ofan alkyl-amino-ethoxy side chain being present (Jordan., 1998). Estrogen and 4-0HT share the same binding site on the ER (Brzozowski et al., 1997). Since they compete for the same binding domain on the ER, 4-0HT was used to test the involvement ofthe ER in alteration ofsynaptic transmission. 4-0HT has a binding affinity 339 greater than that ofestrogen for the binding site on estrogen receptor B(ERB) and 178 times greater for ERa (Kuiper et al., 1997). Preliminary data shows that the addition of4-0HT(60 nM) to the 74 bathing saline was followed by an increase in EPSP amplitude (Fig. 16). This may be due to the displacement ofchemicals previously attached to the estrogen ligand-binding site, suggesting that there may be a certain level of endogenous tonic inhibition. Preliminary data also shows that 4-0HT prevented the inhibition ofexcitatory synaptic activity by estrogen (Fig. 16). This suggests the possibility that ER activation is necessary for the alterations ofsynaptic activity seen in this study. Defining the non-genomic mechanism by which estrogen-induced alterations in synaptic activity are occurring is speculative. Mainly, this is due to the limited knowledge ofER function. Genomic mechanisms of estrogen action are the most clearly understood. However, the genomic mechanism takes hours from estrogen binding and initiation to completion and release ofpost-translationally modified products (Zakon., 1997). In the case ofalteration ofsynaptic activity by estrogen, the period required to see effects (20 -30 minutes) is too short to be a product ofthe genomic mechanism. Alteration ofsynaptic activity by estrogen is most likely occurring via a rapid, non-genomic mechanism. A possible mechanism through which estrogen may be altering synaptic function in the BLA is via various protein kinase pathways. Mitogen activated protein kinase (MAPK), protein kinase A (PKA), and protein kinase C (PKC) are protein kinase 75 pathways that may serve as potential alternative pathways for estrogen function. Studies ofprimary cultured cortical neurons have shown that estrogen induces an increase in MAPK activity within 30 minutes (Singer, et al 1999). Estrogen has also been shown to initiate activation ofMAPK in neuroblastoma cells (Watters et al., 1997), and non-neuronal cells via non receptor kinase src (Migliaccio et al., 1996). Ifthe estrogen receptor antagonist leI 182,780 or a MAPK pathway inhibitor is used in conjunction with estrogen, there is no increased kinase activity in primary cortical neurons (Singer et al., 1999). This implies that estrogen activates MAPK through the activation ofan ER in cultured cortical neurons. MAPK is activated by many intracellular signals, including nerve growth factor (NGF) (Seger and Krebs, 1995). NGF has been suggested to promote survival ofneurons in several cases (Kromer, 1987; Hefti, 1986). Estrogen is also believed to modulate neurotrophins (such as NGF) levels (Gibbs et al., 1994; Singh et al., 1995), and neurotrophin receptors (Sorabji et al., 1994a, b; McMillan et al., 1996). This suggests the possibility that estrogen and neurotrophins may participate in a negative feedback mechanism to control the amount ofkinase activity mediated by the estrogen receptor, NGF, and other factors. 76 Another pathway through which estrogen may be inducing its actions is the protein kinase A (PKA) pathway. In guinea pig hypothalamic slices, estrogen weakens the ability ofJ,!-opioids to hyperpolarize beta-endorphin neurons. This action ofestrogen takes place within 20 minutes, and it was blocked by the ER antagonists ICI 164,384 and diethylstilbestrol. Furthermore, this action was not prevented by the protein synthesis inhibitor cyclohexamide but was blocked by selective PKA antagonists (Lagrange et al., 1997). Another possible pathway for rapid actions ofestrogen in the BLA is by protein kinase C (PKC). In normal and tumor tissue, estrogen has been shown to increase PKC levels (Drouva et al., 1990, Maeda et al., 1993, Maizels et aI., 1993). Although the actions ofthe kinases in relation to estrogen action have not been investigated in the BLA, the demonstration ofrapid (30 min) estrogen action, with inhibition by an ER antagonist, make them plausible mechanisms for the synaptic activity alteration found in BLA neurons, and potential targets for future research. Some ofthe effects ofestrogen occurring in the brain have been shown to be associated with NMDA receptors, due to the recent findings of Woolley and McEwen (1994), who showed that the dendritic spine density 77 changes occurring in the CA1 region ofthe hippocampus following estrogen treatment rely upon the activation ofNMDA receptors. In addition, the competitive NMDA antagonist MK 801 prevents estrogen-induced changes in spine density. Metabotropic and ionotropic NMDA receptor were reportedly affected (Woolley and McEwen 1994). This has implications on our study because BLA neuronal EPSPs rely in part upon the activation of NMDA receptors (Rainnie et al., 1991a). Blocking ofER by tamoxifen shows that alteration ofsynaptic behavior may be dependent upon on a specific activating function (AF) of the ER. AF-l is located in the N terminus and AF-2 is located in the ligand binding domain ofERa. These are two areas suggested to mediate transcriptional activation ofERa (Tsai and O'Malley, 1994). ERa antagonists inhibit and agonists activate AF-2, but some antagonists have been reported to have agonistic actions (Berry et al., 1990). AF-lofERa has been shown to be regulated by growth factors associated with the MAPK pathway, and also may be the cause ofagonistic effects ofER antagonists (McEwen, 1997; Kato et al., 1995). While analyzing the preliminary blocking effect oftamoxifen, it is plausible to hypothesize that the actions ofestrogen seen in BLA pyramidal neurons are mediated by the AF-2 region on the ER. Distribution ofERs in 78 the amygdala (Shughrue et al., 1998; Weiland, 1997) suggests that both ERa and ER~ may be present in the BLA. The ligand binding domains ofERa and ER~ show a 50% homology (Kuiper, et al., 1997), thus the nearby AF-2 regions ofthe ERs may have similar homologies. Therefore, defining which ofthe ER subtypes is involved the alteration ofsynaptic activity in the BLA can help find a more specific location for the actions described here. Passive and intracellu1ar1y stimulated membrane activity recordings did not show significant alteration upon estrogen treatment. Resting membrane potential, early and late membrane resistance, action potential frequency, and action potential accommodation response showed no significant change upon switching the bathing solution to estrogen. This data suggests that the passive currents analyzed in resting membrane potential measurements show no conductance change, and the voltage-gated currents analyzed in the early membrane resistance, late membrane resistance, and action potential accommodation response also show no conductance change. The accommodation response results from the action ofa slowly decaying after hyperpo10rization current (Womble and Moises, 1993). This Ca++-activated K+ current was not significantly changed. Action potential frequency allows us to analyze the voltage-gated Na+ and K+ currents associated with action potential formation (Kandel et al., 1995). 79 These currents had no significant change. Therefore, no changes were found in the Na+ and K+ ion currents, which are known to underlie, active neuronal responses, such as action potential firing patterns (Kandel et aI, 1995). Voltage recordings from BLA neurons presented in this study revealed significant decreases in EPSP amplitude and spontaneous synaptic activity upon estrogen treatment. This is a rapid (20-30 min.) response. 4 OHT appeared to block this effect ofestrogen, therefore suggesting an ER mediated response. The speed ofthe response suggests that the ER is not acting by the classical genomic mechanism ofER action. Voltage recordings analyzing passive and voltage-gated ion channels show no significant change. With the evidence ofprotein kinase pathways associated with estrogen and ER phosphorylation, it would seem likely that some modulation ofion channels should be occurring. Kinases have been found to effect ion channels in past studies ofestrogen action in hippocampal neurons (Gu and Moss, 1996; Kawasaki et aI, 1994; Wyneken et aI, 1997; Potier and Rovira, 1999; Ghetti and Heinemann, 2000). The decreases in EPSP amplitude seen in this study are attributed to changes of the glutamate system. Preliminary evidence showed estrogen does not affect IPSP amplitude. Thus, the GABA system does not appear to be affected. Estrogen does not appear to affect any ofthe membrane properties ofthe 80 BLA neurons studied; therefore, ion channels ofthe studied neurons do not appear to be affected. In conclusion, the accumulated evidence appeared to suggest that estrogen is acting to inhibit the glutamate system. Reduction in glutamate function at BLA synapses supports previous studies that suggest estrogen protects from the glutamate-associated excitotoxicity associated with cerebral ischemia (Hum PD, Macrae, 2000; Sawada et al., 2000; Rusa et ai., 1999; Fung et al., 1999). 81 References Ali S, Metzger D, Bomert JM, Chambon P (1993). Modulation of transcriptional activation by ligand-dependent phosphorylation ofthe human oestrogen receptor AlB region. EMBO Journal. 12:1153. Balthazart J, Ball GF. (1998). New insights into the Regulation and Function ofBrain Estrogen Synthase (aromatase). Trends in Neuroscience. 21:243-249. Barraclough DJ, Ingram CD, Brown MW. (1999). Chronic treatment with oestradiol does not alter in vitro LTP in subfield CAl ofthe female rat hippocampus. Neuropharmacology. Jan;38(1):65-71. Bartus RT, Dean RL, Beer B, and Lippa AS. (1982). The cholinergic hypothesis ofgeriatric memory dysfunction. Science. 217:408-417. Bedard P, Langelier P, Villeneuve A. (1977). Oestrogens and extrapyramidal system. Lance.t Dec 24-31;2(8052-8053):1367-1368 Ben-Ari Y, Zigmond RE, Shute CCD, and Lewis PR. (1977). Regional distribution ofcholine acetyltransferase and acetylcholinesterase within the amygdaloid complex and stria terminalis system. Brain Research. 120:435-445. Berry M, Metzger P, Chambon (1990). Role ofthe two activating domains of the oestrogen receptor in the cell-type and promoter-context dependent agonistic activity ofthe anti-oestrogen 4-hydroxy tamoxifen.. EMBO Journal. 9:2811. Bi R, Broutman G, Foy MR, Thompson RF, Baudry M. (2000). The tyrosine kinase and mitogen-activated protein kinase pathways mediate multiple 82 effects ofestrogen in hippocampus. Proceedings ofthe National Academy ofScience ofthe USA. Mar 28;97(7):3602-3607. Birge SJ, Mortel KF. (1997). Estrogen and the Treatment ofAlzheimer's Disease. American Journal ofMedicine. 103(3A):36S-45S. Black LJ, Jones CD, Falcone JF. (1983). Antagonism ofestrogen action with a new benzothiophene derived antiestrogen. Life Science. 32:1031-1036. Bondi MW and Lange KL. (1998). Alzheimer's Disease. Encyclopedia of Mental Health, Volume 1. San Diego, California: Academic Press. Boyles JK, Notterpek LM, Anderson LJ. (1990). Accumulation of apolipoproteins in the regenerating and remyelinating mammalian peripheral nerve. Journal ofBiological Chemistry. 265:17805-17815. Boyles JK, Zoeliner CD, Anderson LJ, Kosik LM, Pitas RE, Weisgraber KH, Hui DY, Mahley RW, Gebicke-Haerter PH, Ignatius MJ, Shooter EM. (1989). A role for apolipoprotein E, apolipoprotein E, apolipoprotein A 1, and low density lipoprotein receptors in cholesterol transport during regeneration and remyelination ofthe rat sciatic nerve. Journal of Clinical Investigation. 83: 1015-1031. Brenner DE, Kukull WA, Stergachis A, Van Belle G, Bowen JD, McCormick WC, Teri L, Larson EB. (1994). Postmenopausal estrogen replacement therapy and the risk ofAlzheimer's disease: a population based case control study. American Journal ofEpidemiology. 140:262-267. Brzozowski AM, Pike ACW, Dauter Z, Hubbard RE, Bonn T, Engstrom 0, Ohman L, Greene GL, Gustafsson J, Carlquist M. (1997). Molecular Basis ofAgonism and antagonism in the Oestrogen Receptor. Nature. 389: 753-758. Canonaco M, Tavolaro R, Maggi A. (1993). Steroid hormones and receptors ofthe GABAA supramolecular complex. ILProgesterone and estrogen inhibitory effects on the chloride ion channel receptor in different 83 forebrain areas ofthe female rat. Neuroendocrinology. May;57(5):974 84. Carlsen J and Heimer L. (1986). A correlated light and electron microscopic immunocytochemical study ofcholinergic terminals and neurones in the ra1 amygdaloid body with special emphasis on the baso1atera1 amygdaloid nucleus. Journal ofComparative Neurology. 244:121-136. Carlsen J, Zaborsky L, and Heimer L. (1985). Cholinergic projections from the basal forebrain to the baso1atera1 amygdaloid complex: a combined retrograde fluorescent and immuno-histochemica1 study. Journal of Comparative Neurology. 234:155-167. Chao HM, Ma LY, McEwen BS, Sakai RR. (1998). Regulation of Glucocorticoid Receptor and Mineralocorticoid Receptor Messenger Ribonucleic Acids by Selective Agonists in the Rat Hippocampus. Endocrinology. 139:1810-1814. Chao HM, Sakai RR, Ma LY, McEwen BS. (1998). Adrenal Steroid Regulation ofNeurotrophic Factor Expression in the Rat Hippocampus. Endocrinology. 139: 3112-3118. Chao HM, Spencer RL, Frankfurt M, McEwen BS. (1994). The Effects of Aging and hormonal Manipulation on Amyloid Precursor Protein APP695 mRNA expression in the Rat Hippocampus. Journal of Neuroendocrinology. 6:517-521. Chiaia N, Foy M, Tey1er TJ. (1983). The hamster hippocampal slice: II. Neuroendocrine modulation. Behavioral Neuroscience. Dec;97(6):839 843 Clarke WP, Goldfarb J. (1998). Estrogen Enhances a 5-HT1a Response in Hippocampal Slices from Female Rats. European Journal of Pharmacology. 160: 195-197. 84 Connors BW, Gutnick JA, Prince DA. (1982) Electrophysiological properties ofneocortical neurons in vitro. Journal ofNeurophysiology. 48: 1302 1320. Cordoba Montoya DA, Carrer HF. (1977). Estrogen Facilitates Induction of Long Term Potentiation in the Hippocampus ofawake rats. Brain Research. 778:430-438. Cotman CW, Pike CJ, Copani A. (1992). ~-amyloid neurotoxicity: a discussion ofin vitro findings. Neurobiology Aging. 13:587-590. Couse JF, Lindzey J, Grandien K, Gustafsson J, Korach KS. (1997). Tissue Distribution and Quantitative Analysis ofEstrogen Receptor-a (ERa) and Estrogen Receptor-~ (ER~) Messenger Ribonucleic Acid in the Wild-Type and ERa-Knockout Mouse. Endocrinology. 138: 4613-4621. Dodt and Misgeld U. (1986). Muscarinic slow excitation and muscarinic inhibition ofsynaptic transmission in the rat neostriatum. Journal of Physiology. 280:593-608. Drouva SV, Gorenne I, Laplavte E, Rerat E, Enjalbert A, and Kordon C. (1990). Estradiol modulates protein kinase C activity in the rat pituitary in vivo and in vitro. Endocrinology 126, 536-544. Dutar P and Nicoll RA. (1986). Classification ofmuscarinic responses in hippocampus in terms ofreceptor subtypes and second messenger systems electrophysiological studies in vitro. Journal ofNeuroscience. 8:4214-4224. Emson PC, Paninos G, Le Gal La Salle G, Ben Ari Y, and Silver A. (1979). Choline acetyltransferase and acetylcholinesterase containing projections from the basal forebrain to the amygdaloid complex ofthe rat. Brain Research. 165:271-282. Emson PC, Paninos G, Le Val La Salle G, Ben-Ari Y, and Silver A. (1979). Choline acetyltransferase and acetylcholinesterase containing projections 85 from the basal forebrain to the amygdaloid complex ofthe rat. Brain Research. 165:271-282. Feng W, Ribeiro RC, Wagner, RL, Nguyen H, Apriletti JW, Fletterick RJ, Baxter JD, Kushner PJ, West BL. (1998). Hormone-Dependent Coactivator Binding to a Hydrophobic Cleft on Nuclear Receptors. Science. 280: 1747-1749. Foy MR, Teyler TJ, Vardaris RM. (1982). Effects ofdelta 9-THC and 17-P estradiol in hippocampus. Brain Research Bulletin. Apr;8(4):341-345. Foy MR, Teyler TJ. (1983). 17-a-Estradiol and 17-p-estradiol in hippocampus. Brain Research Bulletin. Jun;10(6):735-739. Foy MR, XU J, Xie X, Brinton RD, Thompson RF, Berger TW. (1999). 17P Estradiol Enhances NMDA Receptor-Mediated EPSPs and Long-Term Potentiation. Journal ofNeurophysiology. 81: 925-929. Fung MM, Barrett-Connor E, Bettencourt RR. (1999). Hormone replacement therapy and stroke risk in older women. Journal of Womens Health. 8(3):359-364. Gean PW, Shinnick-Gallagher P (1988) Characterization ofthe epileptiform activity induced by magnesium-free solution in rat amygdala slices: an intracellular study. Experimental Neurology. 101:248-255. Gean PW, Shinnick-Gallagher P (1989) The transient outward current in a mammalian central neurone blocked by 4-aminopyradine. Nature. 299:252-254. Ghetti A, Heinemann SF. (2000). NMDA-Dependent modulation of hippocampal kainate receptors by calcineurin and Ca(2+)/calmodulin dependent protein kinase. Journal ofNeuroscience. Apr 15;20(8):2766 2773. 86 Goodman Y, Bruce AJ, Cheng B, Mattson MP. (1996). Estrogens Attenuate and Corticosterone Exacerbates Excitotoxicity, Oxidative Injury, and Amyloid ~ Peptide Toxicity in Hippocampal Neurons. Journal of Neurochemistry. 66: 1836-1844. Gorczynska E, Handelsman DJ. (1995). Androgens Rapidly Increase the Cytosolic Calcium Concentration in Sertoli Cells. Endocrinology. 136:2052-2059. Gradishar WJ and Jordan VC (1997). Clinical potential ofnew antiestrogens. Journal ofClinical Oncology. 15:840-852. Greco B, Edwards DA, Michael RP, Clancy AN. (1998). Androgen Receptors and Estrogen Receptors are Co-localized in Male Rat Hypothalamic and Limbic Neurons that Express Fos Immunoreactivity Induced by mating. Neuroendocrinology. 67:18-28. Gu Q, Moss RL J. (1996). 17 beta-Estradiol potentiates kainate-induced currents via activation ofthecAMP cascade. Neuroscience. Jun ;16(11):3620-3629. Gu Q, Moss RL. (1996). 17~-Estradiol Potentiates Kainate-Induced Currents via Activation ofthe cAMP Cascade. Journal ofNeuroscience. 16(11):3620-3629. Gu Q, Moss RL. (1998). Novel mechanism for non-genomic action of17 beta oestradiol on kainate-induced currents in isolated rat CA1 hippocampal neurones. Journal Physiology (London). Feb 1;506:745-754 Haas HL. (1982). Cholinergic disinhibition in hippocampal slices ofthe rat. Brain Research. 233:200-204. Hefti T. (1986). Nerve Growth Factor Promotes Survival ofSeptal Cholinergic Neurons After Fimbrial Transections. Journal ofNeuroscience. 6(8):2155-2162. 87 Hellendall RP, Godfrey DA, Ross CD, Armstrong DM, and Prive JL. (1996). The distribution ofcholine acetyltransferase in the rat amygdaloid complex and adjacent cortical areas as determined by quantitative micro assay and immunohistochemistry. Journal ofComparative Neurology. 249:486-498. Henderson Y W. (1997). Estrogen, Cognition, and a Woman's Risk of Alzheimer's Disease. American Journal ofMedicine. 103(3A): 11 S-18S. Hill CS, and Treisman R. (1995). Transcriptional regulation by extracellular signals: mechanisms and specificity.. Cell. 80: 199-211. Holtzman DM, Pitas RE, Kilbridge J, Nathan B, Mahley RW, Bu G, Schwartz AL. (1995). Low density lipoprotein receptor related protein mediates apolipoprotein E-dependent neurite outgrowth in a central nervous system derived neuronal cell line. Neurobiology. 92:9480-9484. Hum PD, Macrae 1M Estrogen as a neuroprotectant in stroke. (2000). J Cerebral Blood Flow and Metabolism.. 20(4):631-652. Jaffe AB, Toran-Allerand CD, Greengard P, Gandy SE. (1994). Estrogen Regulates Metabolism ofAlzheimer Amyloid ~ Precursor Protein. The Journal ofBiochemistry andMolecular Biology. 269: 13065-13068, 1994. Jensen EY, Mohla S, Brecher PI, DeSombre ER, (1973). Estrogen receptor transformation and nuclear RNA synthesis. Advanced Experimental Medical Biology. 36:60-79. Johnson MG and Everitt BJ (1995). Essential Reproduction, Fourth Edition. Cambridge, Massachusetts: Blackwell Science. Jordan YC. (1984). Biochemical pharmacology ofantiestrogen action. Pharmacology Reviews. 36:245-256. 88 Jordan VC. (1998). Antiestrogenic Action ofRaloxifene and Tamoxifen: Today and Tomorrow. Journal ofthe National Cancer Institute. 90: 967-971. Kandel ER, Schwartz JH, Jessell TM (1995). Essentials ofNeural Science and Behavior. Norwalk, Connecticut: Appleton and Lange. Kato S, Endoh H, Masuhiro Y, Kitamoto T, Uchiyama s, Sasaki H, Masushige S, Gotoh Y, Nishida E, Kawashima H, Metzger D, Chambon P. (1995). Activation ofthe Estrogen Receptor Through Phosphorylation by Mitogen-Activated Protein Kinase. Science. 270:1491-1494. Katzenellenbogen JA, O'Malley BW, and Katzenellenbogen BS (1996). Molecular Endocrinology. 10: 119-131. Kawas C, Resnick S, Morrison A, Brookmeyer R, Corrada M, Zonderman A, Baca1 C,Lingle DD, Metter E. (1997). A prospective study ofestrogen replacement therapy and the risk ofdevelopingAlzheimer's disease: the Baltimore Longitudinal Study ofAging. Neurology. Jun;48(6):1517-21 Kawasaki M, Uchida S, Monkawa T, Miyawaki A, Mikoshiba K, Mammo F, Sasaki S. (1994). Protein, Nucleotide Cloning and expression ofa protein kinase C-regu1ated chloride channel abundantly expressed in rat brain neuronal cells. Neuron. Mar;12(3):597-604. Korach KS. (1994). Insights from the Study ofAnimals Lacking Functional Estrogen Receptor. Science. 266: 1524-1527. Kow LM, PfaffDW. (1984). Suprachiasmatic neurons in tissue slices from ovariectomized rats: e1ectrophysio1ogical and neuropharmacological characterization and the effects ofestrogen treatment. Brain Research. Apr 16;297(2):275-286. Kromer LF. (1987). Nerve Growth Factor Treatment After Brain Injury Prevents Neuronal Death. Science. 235:214-216. 89 Kubo K, Gorski RA, Kawakami M. (1975). Effects ofestrogen on neuronal excitability in the hippocampa1-septa1-hypotha1amic system. Neuroendocrinology. 18(2):176-91. Kuiper G, Carlsson B, Grandien K, Enmark E, Haggb1ad J, Nilsson S, Gustafsson J. (1997). Comparison ofthe Ligand Binding Specificity and Transcript Tissue Distribution ofEstrogen Receptors a and p. Endocrinology. 138: 863-870. Kumar V, Green S, Stack G, Berry M, Jin J, Chambon P. (1987). Functional Domains ofthe Human Estrogen Receptor. Cell. 51: 941-951. Lagrange AH, Ronnek1eiv OK, Kelly MJ. (1994). The Potency ofJl-Opioid Hyperpolarization ofHypothalamic Arcuate Neurons is Rapidly Attenuated by 17B-Estradiol. The Journal ofNeuroscience. 14(10): 6196-6204. Lagrange AH, Ronnek1eiv OK, Kelly MJ. (1997). Modulation ofG Protein Coupled Receptors by an Estrogen Receptor that Activates Protein Kinase A. Molecular Pharmacology. 51: 605-612. Li X, Schwartz PE, Rissman EF. (1997). Distribution ofEstrogen Receptor B like Immunoreactivity in Rat Forebrain. Neuroendocrinology. 66:63-67. Madison DV, Nicoll RA. (1984). Control ofthe repetitive discharge ofrat CAl pyramidal neurons in vitro. Journal ofPhysiology (London). 354:319-331. Maeda T, and L10ud RV. (1993) Protein kinase C activity and messenger RNA modulation by estrogen in normal and neoplastic rat pituitary tissue.. Laboratory Investigations. 68,472-480. Mah1ey RW (1988). Apo1ipoprotein E: cholesterol transport role with expanding role in cell biology. Science. 240:622-630. 90 Maizels, E. T., Miller, J. B., Cutler, f. J., Jackiw, V., Carney, E. M., Mizuno, K., 0000, S., and Hunzicker, D. M. (1992). Journal ofBiological Chemistry. 267, 17061-17068. Martin JH (1996). Neuroanatomy: Text andAtlas, Second Edition. Stamford, Connecticut: Appleton and Lange. Mason RP, Shoemaker WJ, Shajenko L, Chambers TE, Herbette LG. (1991). Evidence for changes in the Alzheimer's disease brain cortical membrane structure mediated by cholesterol. Neurobiology ofAging. 13:143-419. Maury APJ. (1995). Biology ofdisease: molecular pathogenesis ofp amyloidoses. Laboratory Investigation. 72:4-16. Mayeux R, Ottman R, Maestre G, Ngai C, Tang MX, Ginsberg H, Chun M, Tycko B, Shelanski M.. (1995). Synergistic effects oftraumatic head injury and apolipoprotein-epsilon 4 in patients with Alzheimer's disease. Neurology. 45:555-557. McCormick DA, Connors BW, Lightahall JW, Prince DA (1985). Comparative electrophysiology ofpyramidal and sparsely spiny stellate neurons ofthe neocortex. Journal ofNeurophysiology. 54:782-806. McCormick DA, Connors BW, Lighthall JW, Prince DA. (1985). Comparative electrophysiology ofpyramidal and sparsely spiny stellate neurons ofthe neocortex. Journal ofNeurophysiology. 54:782-806. McDonald AJ (1982). Neurones ofthe lateral and basolateral amygdaloid nuclei: a Golgi study in the rat. Journal ofComparative Neurology. 212:293-312. McDonald AJ (1984). Neuronal organization ofthe lateral and basolateral amygdaloid nuclei in the rat. Journal ofComparative Neurology. 222:589-606. 91 McEwen BS, Alves SE, Bullock K, Weiland NG. (1997). Ovarian Steroids and the Brain: Implications for cognition and aging. Neurology. 48:S8 S15. McEwen BS, Tanapat P, Weiland NG. (1997). Inhibition ofDendritic Spine Induction on Hippocampal CAl Pyramidal Neurons by a Non-steroidal Estrogen Antagonist in Female Rats. Endocrinology. 140(3):1044-1047. Metzger D, Berry M, Ali S, Chambon (1995). Effect ofantagonists on DNA binding properties ofthe human estrogen receptor in vitro and in vivo. Molecular Endocrinology. 9:579-591. Millhouse OE and DeOlmos J (1983). Neuronal configuration in lateral and basolateral amygdala. Neuroscience. 10: 1269-1300. Murphy DD, Segal M (1996). Regulation ofdendritic spine density in cultured rat hippocampal neurons by steroid hormones. Journal ofNeuroscience. 16:4059-4068. Ohkura T, Isse K, Akazawa K, Hamamoto M, Yaoi Y, Hagino N. (1994). Related Articles Evaluation ofestrogen treatment in female patients with dementia ofthe Alzheimer type. Journal ofEndocrinology. 41(4):361 371. Paxinos GT, Watson C. (1997). The Rat Brain in Stereotaxic Coordinates. Academic Press, Incorporated. Pelech SL and Sanghera (1992). MAP kinases: charting the regulatory pathways.. Science. 257: 1355-1356. Pike CJ, Walencewicz AJ, Glabe CG, Cotman CWo (1991). In vitro aging of ~-amyloid protein causes peptide aggregation and neurotoxicity. Brain Research. 563:311-314. 92 Poirier J, Hess M, May PC, Finch CE (1991). Astrocytic apolipoprotein E mRNA and GFAP mRNA in hippocampus after entorhinal cortex lesioning. Molecular Brain Research. 11:97-106. Potier B and Rovira C. (1999). Related Protein tyrosine kinase inhibitors reduce high-voltage activating calcium currents in CA1 pyramidal neurones from rat hippocampal slices. Brain Research. Jan 23;816(2):587-597. Puy L, MacLusky NJ, Becher L, Karsan N, Trachtenberg J, Brown TJ. (1995). Immunocytochemical Detection ofAndrogen Receptor in Human Temporal Cortex: Characterization and Application ofPolyclonal Androgen Receptor Antibodies in Frozen and Paraffin-embedded Tissues. Journal ofSteroid Biochemistry and Molecular Biology. 55:197-209. Rainnie DG Asprodini EK, Shinnick-Gallagher P (1991a). Excitatory transmission in the basolateral amygdala. Journal ofNeurophysiology. 66:986-998. Rainnie DG Asprodini EK, Shinnick-Gallagher P (1991b). Inhibitory transmission in the basolateral amygdala. Journal ofNeurophysiology. 66:999-1009. Recasens M, Guiramand J, Vignes M. (1991). The putative molecular mechanism(s) responsible for the enhanced inositol phosphate synthesis by excitatory amino acids: an overview. Neurochemistry Research. 16:659-668. Reibel S, Andre V, Chassagnon S, Andre G, Marescaux C, Nehlig A, Depaulis A. (2000). Neuroprotective effects ofchronic estradiol benzoate treatment on hippocampal cell loss induced by status epilepticus in the female rat. Neuroscience Letters. Mar 10;281(2-3):79-82. 93 Ripich DN, Petrill SA, Whitehouse PJ, Ziol EW. (1995). Gender Differences in Language ofAD Patients: A Longitudinal Study. Neurology. 45: 299 302. Rusa R, Alkayed NJ, Crain BJ, Traystman RJ, Kimes AS, London ED, Klaus JA, Hum PD. (1999). 17~-estradiol reduces stroke injury in estrogen deficient female animals. Stroke. 30(8):1665-1670. SanMartin S, Gutierrez M, Menendez L, Hidalgo A, Baamonde A. (1999). Effects ofdiethylstilbestrol on mouse hippocampal evoked potentials in vitro. Neurobiology. 19(6):691-703. Sawada M, A1kayed NJ, Goto S, Crain BJ, Traystman RJ, Shaivitz A, Nelson RJ, Hum PD. (2000). Estrogen receptor antagonist ICI182,780 exacerbates ischemic injury in female mouse. Journal Cerebral Blood Flow andMetabolism. Jan;20(1):112-118. Shiau AK, Barstad D, Loria PM, Cheng L, Kushner PJ, Agard DA, Greene GL. (1998). The Structural Basis ofEstrogen Receptor/Coactivator Recognition and the Antagonism ofthis Interaction by Tamoxifen. Cell. 95:927-937. Shughrue PJ, Scrimo PJ, Merchenthaler. (1998). Evidence for the Co localization ofestrogen Receptor B mRNA and Estrogen Receptor a Immunoreactivity in Neurons ofthe Rat Forebrain. Endocrinology. 139(12): 5267-5270. Singer CA, Figueroa-Masot XA, Batchelor RH, Dorsa DM. (1999). The Mitogen-activated Protein Kinase Pathway Mediates Estrogen Neuroprotection after Glutamate Toxicity in Primary Cortical Neurons. Journal ofNeuroscience. 19(7):2455-2463. Smigel K (1998). Breast cancer prevention trial shows major benefit some risk. Journal ofthe National Cancer Institute. 90:647-648. 94 Smith SS, Water house BD, Woodward DJ. (1987). Sex steroid effects on extrahypothalamic CNS. 1. Estrogen augments neuronal responsiveness to iontophoretically applied glutamate in the cerebellum. Brain Research. 422: 50-51. Smith SS. (1989). Estrogen administration increases neuronal responses to excitatory amino acids as a long-term effect. Brain Research. 503:354 357. Teyler TJ, Vardaris RM, Lewis D, Rawitch AB. (1980). Gonadal steroids: effects on excitability ofhippocampal pyramidal cells. Science. Aug 29;209(4460): 1017-8. Toran-Allerand CD, Miranda RC, Bentham W, Sohrabji F, Brown TJ, Hochberg RB, MacLusky NJ. (1992). Estrogen Receptors Colocalize with Low-Affinity Nerve Growth Factor Receptors in Cholinergic Neurons ofthe Basal Forebrain. Neurobiology. 89: 4668-4672. Tsai, M and O'Malley BW. (1994). Molecular Mechanisms ofAction of Steroid/Thyroid Receptor Superfamily Members. Annual Review of Biochemistry. 63 :451-486. Valentino, RJ, and Dingledine R. (1981). Presynaptic inhibitory effect of acetylcholine in the hippocampus. Journal ofNeuroscience. 1:784-792. Vardaris RM, Lewis D, Rawitch AB. (1990) Gonadal steroids: effects on excitability ofhippocampal pyramidal cells. Science. 209: 1017-1019. Washburn MS, Moises HC (1992a) Inhibitory responses ofrat basolateral amygdaloid neurons recorded in vitro. Neuroscience. 50:811-830. Washburn MS, Moises HC. (1992b) Electrophysiological and Morphological Properties ofRat Basolateral Amygdaloid Neurons in vitro. The Journal ofNeuroscience. 12(10): 4066-4079. 95 Washburn MS, Moises HC. (1992c) Muscarinic Responses ofRat Basolateral Amygdaloid Neurons Recorded in vitro. Journal ofPhysiology. 449: 121-154. Weiland NG, Orikasa C, Hayashi S, McEwen BS. (1997). Distribution and Hormone Regulation ofEstrogen Receptor Immunoreactive Cells in the Hippocampus ofMale and Female Rats. The Journal ofComparative Neurology. 388:603-612. Weiland NG. (1992). Estradiol Selectively Regulates Agonist Binding Sits on the N-Methyl-D-Aspartate Receptor Complex in the CAl Region ofthe Hippocampus. Endocrinology. Vol. 131, no. 2: 662-668. Womble MD, Moises HC (1992). Voltage-clamp analysis ofcholinergic action in the basolateral amygdala. Journal ofPhysiology. 457:93-114. Womble MD, Moises HC. (1993). Muscarinic modulation ofconductances underlying the afterhyperpolarization in neurons ofthe rat basolateral amygdala. Brain Research. Sep 3;621(1):87-96. Wong M, Moss RL. (1991). Electrophysiological evidence for a rapid membrane action ofthe gonadal steroid, 17 beta-estradiol, on CAl pyramidal neurons ofthe rat hippocampus. Brain Research. Mar 8;543(1):148-152. Wong M, Moss RL. (1992). Long-term and short-term electrophysiological effects ofestrogen on the synaptic properties ofhippocampal CAl neurons.. Journal ofNeuroscience. 12(8):3217-3225. Wong M, Moss RL. (1992). Modulation ofsingle-unit activity in the rat medial amygdala by neurotransmitters, estrogen priming, and synaptic inputs from the hypothalamus and midbrain. Synapse. Feb;10(2):94 102. Wong RK, Prince DA (1981). Afterpotential generation in hippocampal pyramidal cells. Journal ofNeurophysiology. 45:86-97. 96 WoolfNJ and Butcher LL. (1982). Cholinergic projections to the basolateral amygdala: a combined Evans Blue and acetylcholinesterase analysis. Brain Research Bulletin. 8:751-763. Woolley CS, McEwen BS. (1994). Estradiol Regulates Hippocampal Dendritic Spine Density via an N-Methyl-D-Aspartate Receptor-Dependent Mechanism. Journal ofNeuroscience. 14(12): 7680-7687. Woolley CS, Weiland NG, McEwen BS, Schwartzkroin PA. (1997). Estradiol increases the sensitivity ofhippocampal CAl pyramidal cells to NMDA receptor-mediated synaptic input: correlation with dendritic spine density. Journal ofNeuroscience. Mar 1;17(5):1848-1859. Woolley CS. (1998). Estrogen-mediated structural and functional synaptic plasticity in the female rat hippocampus. Hormones and Behavior. Oct;34(2):140-148. Woolley CS. (1999). Effects ofEstrogen in the CNS. Current Opinion in Neurobiology. 9: 349-354. Wrenn CK, Katzenellenbogen BS. (1993). Structure-Function Analysis ofthe hormone Binding Domain ofthe Human Estrogen Receptor by Region specific Autogenesis and Phenotypic Screening in Yeast. The Journal of Biochemistry andMolecular Biology. 268: 24089-24098. Wyneken U, Riquelme G, Villanueva S, Orrego F. (1997). Effect ofglutamate receptor phosphorylation by endogenous protein kinases on electrical activity ofisolated postsynaptic densities ofcortex and hippocampus. Neuroscience Letters. Mar4;224(2):131-135. Zakon Harold H. (1998) The Effects ofSteroid Hormones on the Electrical Activity ofExcitable Cells. Trends in Neuroscience. 21:202-207. 97