Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2010 Nov 1.
Published in final edited form as: CNS Neurol Disord Drug Targets. 2010 Nov 1;9(5):651–660. doi: 10.2174/187152710793361559

Gonadotropin-releasing hormone receptor system: modulatory role in aging and neurodegeneration

Liyun Wang 1, Wayne Chadwick 1, Soo-Sung Park 1, Yu Zhou 1, Nathan Silver 1, Bronwen Martin 2, Stuart Maudsley 1,*
PMCID: PMC2967575  NIHMSID: NIHMS226428  PMID: 20632963

Abstract

Receptors for hormones of the hypothalamic-pituitary-gonadal axis are expressed throughout the brain. Age-related decline in gonadal reproductive hormones cause imbalances of this axis and many hormones in this axis have been functionally linked to neurodegenerative pathophysiology. Gonadotropin-releasing hormone (GnRH) plays a vital role in both central and peripheral reproductive regulation. GnRH has historically been known as a pituitary hormone; however, in the past few years, interest has been raised in GnRH actions at non-pituitary peripheral targets. GnRH ligands and receptors are found throughout the brain where they may act to control multiple higher functions such as learning and memory function and feeding behavior. The actions of GnRH in mammals are mediated by the activation of a unique rhodopsin-like G protein-coupled receptor that does not possess a cytoplasmic carboxyl terminal sequence. Activation of this receptor appears to mediate a wide variety of signaling mechanisms that show diversity in different tissues. Epidemiological support for a role of GnRH in central functions is evidenced by a reduction in neurodegenerative disease after GnRH agonist therapy. It has previously been considered that these effects were not via direct GnRH action in the brain, however recent data has pointed to a direct central action of these ligands outside the pituitary. We have therefore summarized the evidence supporting a central direct role of GnRH ligands and receptors in controlling central nervous physiology and pathophysiology.

Keywords: Gonadotropin releasing hormone, hypothalamic-pituitary-gonadal axis, brain, GnRH receptors, Alzheimer’s disease, amyloid precursor protein, neuron

INTRODUCTION

Hypothalamic gonadotropin-releasing hormone (GnRH) is a decapeptide that plays a crucial role in the regulation of reproduction as well as controlling many other functions related to reproductive activity outside the hypothalamic-pituitary gonadal (HPG) axis [1]. GnRH is released in synchronized pulses from nerve endings into the hypophyseal portal system every 30–120 minutes to stimulate the biosynthesis and secretion of luteinizing hormone (LH) and follicle-stimulating hormone (FSH) from pituitary gonadotropes. GnRH-I (pGlu-His- Trp-Ser-Tyr-Gly-Leu-Arg-Pro-Gly-NH2) was the first GnRH isoform discovered in the mammalian brain. At least two, and usually three, forms of GnRH are present in most vertebrate species. Amongst these, is a form originally isolated from the chicken, i.e. chicken GnRH II (pGlu-His-Trp-Ser-His-Gly-Trp-Tyr-Pro-Gly-NH2) that was found to be universally present and uniquely conserved between from jawed fish to humans [26]. With respect to the effects of GnRH and its analogues upon the central nervous system (CNS), considerable evidence has accumulated for a role of LH in promoting alterations in neurophysiological activity. For example, epidemiological support for a role of LH/GnRH in Alzheimer’s disease (AD) is evidenced by a reduction in neurodegenerative disease among prostate cancer patients receiving GnRH agonistic therapy [7,8]. We will discuss the evidence supporting the role of the GnRH hormonal receptor system in modulating the biochemical, pathological, and cognitive changes associated with aging and age-related neurodegenerative disorders. An examination of the potential widespread actions that this traditional reproductive hormone exerts may lead to the generation of novel therapies and provide fresh insight into the therapeutic tackling of complex disorders that affect multiple aspects of peripheral and central nervous tissue function.

HYPOTHALAMIC-PITUITARY-GONADAL AXIS IN AGING-RELATED PHYSIOLOGY

In humans, aging is inexorable and is the eventual summation of multiple interacting physiological or pathophysiological effects. “Aging” and the related word “senescence” are commonly used to refer to post-maturational processes that lead to diminished homeostasis and increased organismic vulnerability. This is typically associated with a progressive decline in physical and cognitive functions. There are at least five notable supporting common characteristics of aging in mammals: increased mortality with age after maturation; changes in biochemical composition in tissues with age; progressive decrease in physiological capacity with age; reduced ability to respond adaptively to environmental stimuli with age and an increased susceptibility and vulnerability to disease [9]. Age-related disorders can be separated into those that develop through normal physiological processes (and are often universally presented to some extent in aged populations), e.g. menopause or the decline in renal function, and those that are associated with age-related pathophysiological aging, e.g. AD. Alzheimer’s disease is an example of pathophysiological aging and is therefore not universal to all elderly people. This approach to aging can utilize a conceptual framework that identifies intrinsic (developmental-genetic) versus extrinsic (stochastic) causes. Accumulating evidence increasingly stresses the impact of age-dependent endocrine changes on the dynamics of neuronal behavior, neurodegeneration, cognition, biological rhythms, sexual behavior, and metabolism. Disrupted metabolic homeostasis in the elderly is likely exacerbated by age-dependent molecular alterations in components of signal transduction pathways and declining production of sex steroids, which determines the response to exogenous influences and thereby increases the predisposition to illness and death. Attempts at understanding the causes of aging have been limited by its complexity. However, considerable evidence has suggested that reproductive activity is a major pacemaker of aging and death. A putative integrative site for these regulatory actions is the neurosecretory system, particularly the HPG axis [10,11]. In humans, reproduction is controlled primarily by the HPG axis hormones, found in both central and peripheral locations. Classically, the prime source of GnRH is considered to be the hypothalamus, where it is released into the hypophyseal portal system to activate GnRH receptors on pituitary gonadotropes, to facilitate the synthesis and release of LH and FSH. GnRH can also be produced and act peripherally in many reproductive tissues (prostate, breast, gonads) and non-reproductive tissues (pancreas, immune system) [1214]. These peripheral organs, and especially the gonads, also generate important HPG feedback hormones such as the sex steroids themselves, inhibins, activins and follistatin. The levels of each of these hormones are regulated by multiple complex feedback loops between the gonads, and the hypothalamus and anterior pituitary [15].

The HPG axis undergoes a number of changes throughout the reproductive lifecycle that are responsible for the development, pubertal changes, and eventual senescence of reproductive systems. While the ligand members of the HPG axis are the direct regulators of reproduction, each member of the axis is also likely impacted by other hormonal factors that would be altered under adverse or favorable reproductive and aging conditions. During this natural progression, levels of HPG axis hormones and reproductive tissue activity can be modulated by higher neuronal activities, and in turn also affect these neural networks via feedback loops. Age-related declines in reproductive function results in an imbalance of this hormonal axis, and lead to menopause- and andropause-related pathophysiology in the central nervous system (CNS) and periphery. Eventually, reductions in gonadal sex steroid production lead to a disruption of hypothalamic feedback inhibition, disturbing normal GnRH and gonadotropin production. In women, the loss of this negative feedback by estrogen and inhibins [16] results in significant and long-lasting increases in serum LH and FSH levels [17,18]. Post-menopause gonadotropin concentrations eventually decline but never decrease to levels seen during the reproductive period. Men experience a more gradual and minimal loss of reproductive function, with a corresponding progressive increase in gonadotropin levels. This ultimately leads to a greater than 2- and 3-fold increase in LH and FSH levels, respectively [19]. Following reproductive life, menopause/andropause-mediated changes in serum and neuronal concentrations of HPG axis hormones may significantly alter neuronal signaling mechanisms. It is difficult to ascribe structural and functional changes during development, adulthood and senescence to a single HPG hormone, but the imbalance effects of this altered neuronal signaling on the structure and function of the brain is likely to be connected the development of pathophysiology in multiple neurodegenerative disorders.

DISRUPTION OF HYPOTHALAMIC-PITUITARY-GONADAL AXIS HORMONES IN THE CENTRAL NERVOUS SYSTEM

Epidemiological and biochemical studies have indicated an association between hormones of the HPG axis and cognitive senescence. For example, changes in HPG hormones following menopause/andropause are involved in the cognitive and neuropathological changes observed in familial AD. Aging-mediated increases in neuronal LH have been associated temporally with the increase of neuronal populations at risk of degeneration and death. Elevations in LH parallel the ectopic expression of cell cycle alteration and oxidative markers, which can precede neuronal degeneration by decades [20,21]. LH also can promote reactivation of mitotic signaling pathways shown to occur early in Alzheimer’s pathogenesis [2224]. The isolation of several forms of GnRH in neural tissue of tunicates and their activation of the gonads [25,26], suggests that direct regulation of the gonads evolved before the development of a neuroendocrine role in mammals regulating both the pituitary and gonads. Neurons are probably one of the earliest cells in evolution to synthesize and secrete GnRH peptides. Interestingly, the major CNS biochemical and neuropathologic changes reported for AD, e.g. alterations in Amyloid Precursor Protein (APP) metabolism, Amyloid-β (Aβ) deposition, tau phosphorylation, mitochondrial alterations, chromosomal replication, synapse loss, death of differentiated neurons, may all be the combined result of increased mitotic signaling by gonadotropins and GnRH, decreased differentiative and neuroprotective signaling via sex steroids, and increased differentiation signaling via activins.

Multiple cell surface receptors for hormones of the HPG axis, including the GnRH receptor, that regulate reproductive function are expressed throughout the brain, and in particular the limbic system and hippocampus, two sets of neurons which are vulnerable to AD pathology. Changes in receptor expression and concentration of peripheral circulating hormones related to aging affect their signaling transduction, as well as neuronal structure and function. Hippocampal GnRH receptor expression has been shown to be increased in old rats [27], and also after castration, which partially mimics age-dependent reproductive decline [28]. LH is known to cross the blood-brain barrier and LH receptors are expressed in the brain with the highest expression in brain regions susceptible to AD neuropathology [23]. Additionally, aging also leads to a reduction in hippocampal estrogen or androgen receptor expression in mice and rats [2932]. Evidence supporting a role for gonadotropins in the etiology of AD includes the two-fold elevation in serum gonadotropin concentrations in AD patients, compared to age-matched control subjects [33, 34]. Epidemiologic studies indicating a female predominance of the disease (2:1, female:male) are also consistent with the earlier loss of reproductive function, and an earlier increase in serum gonadotropin concentrations in women [3537]. Based on these epidemiological and biochemical studies, steroidal hormone-replacement therapy was, until recently, viewed as a major factor in the prevention of AD. However, a recent randomized clinical trial revealed that estrogen hormone replacement therapy may actually exacerbate the incidence of dementia, when administered to elderly women [38]. These contradictory reports have cast doubt on the role of estrogen in disease pathogenesis and led us to consider an alternate hypothesis that would be consistent with both observations. Specifically, we suspect that hormones of the hypothalamic pituitary gonadal axis, such as gonadotropins, as well as GnRH, are involved in the pathogenesis of AD. For example, LH is significantly elevated in both the sera and brain tissue of patients with AD and leads to an increased production of Aβ, one of the toxicity-inducing factors of AD. Distribution and expression levels of neuronal receptors for LH corresponds to the populations of neurons that degenerate during the course of this disorder. Evidence that pathogenesis can be mediated outside of gonadal steroids has led to a therapeutic shift to GnRH-based therapies, not only for the treatment of AD, but also for a wide variety of other aging-related disorders.

GnRH and GnRH RECEPTORS IN THE CENTRAL NERVOUS SYSTEM

GnRH neurons are distributed in a loose array along the ventral medial forebrain from the posterior olfactory bulbs to the arcuate nucleus. GnRH neurons are found in the vicinity of the olfactory placode during prenatal development, after which GnRH-producing cells migrate through the nasal system into the forebrain [1, 39, 40]. Therefore, differences in GnRH-producing areas within the brain appear to result from greater or lesser penetration along the olfactory-forebrain-hypothalamus continuum to the median eminence. The GnRH family currently includes 25 isoforms, 14 and 11 from representative vertebrate and invertebrate species, respectively [41]. To date, three forms of GnRH (GnRH I, II and III) has been identified in many vertebrates (e.g. bony fish and amphibians), but in reptiles, birds and mammals only GnRH I and II are apparently present in addition to a truncated form of GnRH I [42]. Therefore, all vertebrates possess a functional second major ligand in the GnRH system, also known as chicken GnRH-II (pGlu-His-Trp-Ser-His-Gly-Trp-Tyr-Pro-Gly-NH2), due to its initial species isolation. As we have mentioned previously, the primary amino acid sequence of GnRH-II has remained unchanged from cartilaginous fish through mammals, which would suggest an immensely strong selective pressure for its conservance. Surprisingly however, its specific molecular physiological role in the CNS is currently poorly understood [41, 43, 44]. The expression pattern of GnRH-II varies from species to species, but is usually highly expressed in the brain and then to a lesser extent in numerous peripheral reproductive and non-reproductive tissues [41, 45]. In humans and primates, GnRH-II is particularly abundant in the caudate nucleus, hippocampus and amygdala [46]. GnRH-II expression in extrahypothalamic regions is often not coincident with expression of the GnRH-I peptide, suggesting unique regulation and functional activities for this conserved peptide. As a caveat to this assumption, there are species though in which GnRH-II is found in the hypothalamic regions and acts at the pituitary for stimulating gonadotropin function [47]. It has been repeatedly proposed however that GnRH II has a neurotransmitter-neuromodulatory role, based on the wide distribution of GnRH II in the central and peripheral nervous system [48, 49].

The actions of GnRH in mammals are mediated by stimulation of the Type I GnRH receptor (GnRHR), which belongs the rhodopsin-like G protein-coupled receptor (GPCR) superfamily. In earlier-developed species, such as sea-squirts, up to three different GnRH receptor isoforms exist. This diversity progressively reduces with more recently evolved animals, even those very closely related in time. For example, humans possess only one functional type of GnRH receptor (Type I) while small pro-simians, such as marmosets, possess two functional GnRH receptors (Type I and Type II receptors). The mammalian Type I GnRH receptor is unique among rhodopsin-like GPCRs, as it has a relatively short third intracellular loop, and lacks an intracellular carboxyl terminus (C-terminus). This C-terminus deletion facilitates the pre-ovulatory LH surge in mammalian reproduction and prevents rapid agonist-induced receptor internalization [50, 51]. In rhodopsin-like GPCRs, this C-terminus typically acts as a substrate for G protein-coupled receptor kinase phosphorylation that initiates the desensitization process of generic rhodopsin-like GPCR, such as the beta-adrenoceptor. GnRH-mediated activation of the Type I GnRHR typically induces Gαq/G11 GDP-GTP exchange, which then stimulates increased phosphoinositol turnover by activating phospholipase C (PLC). This enzyme leads to the generation of several second messengers [52,53], among these, diacylglycerol (DAG) and inositol 1, 4, 5-tris-phosphate (IP3) are critically important. DAG leads to activation of protein kinase C (PKC), and IP3 releases Ca2+ from intracellular pools [54]. Both events, in the anterior pituitary gonadotrope cells, result in gonadotropin synthesis and release [55, 56]. In recent years the true range of GnRH signal transduction pathways that are elicited upon receptor activation has been dramatically expanded. Ligand activation has been shown to mediate: increases in cAMP [57], activation of multiple members of the mitogen-activated protein kinase (MAPK) family [48, 58], inhibition of glycogen synthase kinase-3 [59], stimulation of the non-receptor tyrosine kinases including c-Src [60]and proline-rich tyrosine kinase 2 (Pyk2) [61], and also diacylglycerol kinases [62].

The presence of GnRH II in all vertebrates initially suggested the probable existence of cognate Type II GnRH receptors. This was supported by the presence of GnRH receptors in amphibian sympathetic ganglia that were selective for GnRH II [63]. It was thought that only non-mammalian GnRHRs possessed functional C-termini until the discovery of a mammalian Type II GnRH receptor with a functional C-terminus [48]. However, as yet, a human Type II receptor that selectively responds to GnRH II has not been cloned. A human type II receptor pseudogene homolog carries a frame-shift and a premature stop codon [48,64]. It is likely that the function of the non-mammalian Type II GnRH receptors has become subserved by the Type I receptor in humans thus creating the potential for differential ligand activation of this GPCR [58]. The expression of the Type I GnRHR in multiple cellular contexts has, in-part, revealed that signaling diversity for GnRH I at the Type I receptor can be created.

COMPLEX SIGNALING PATTERNS THROUGH THE GnRH RECEPTORS

With respect to the signaling output from GnRH receptors, it is clear that at the level of the G protein the Type I receptor can interact with multiple G-proteins such as Gαq/11, Gαi, and Gαs in order to mediate various biological actions. A ‘cycle of signaling’ that may decode a GnRH pulse can already be detected at the G-protein level. GnRH increases the mRNA levels of RGS2 (regulators of G-protein signaling), a large family of proteins that modulate G-protein activity by interacting with active Gα-subunits to accelerate their intrinsic GTPase activity and limit their half-life [65]. On the other hand, GnRH activates PKC, which can then phosphorylate RGS2 leading to its inhibition and hence may reduce the deactivation by RGS2 of the Gα-subunits. In addition to interaction with multiple G proteins, the Type I GnRHR can also interact functionally with other receptor systems such as the androgen receptor [66] and in a manner reminiscent of the prototypic GPCR, the β2-adrenoceptor, with the epidermal growth factor receptor [6770]. Indicative of the pluripotent signaling nature of the Type I GnRHR, GnRH receptor stimulation may mediate different, even opposite, responses depending the expression context of the receptor, as well as the agonistic stimulation mode, e.g.. signal responses can be qualitatively diverse at different doses. It has been shown that pulsatile GnRH stimulates more sustained extracellular signal-regulated kinase (ERK) activity (more than 8h), whereas continuous infusion of gonadotrope αT3-1 cells with GnRH stimulates short-term (2h) ERK activity [71]. Also, GnRH treatment can stimulate cAMP production at nanomolar concentrations, but has an inhibitory effect at micromolar concentrations [72]. It should be pointed out that the nanomolar concentration range (0.01–1nM) corresponds to the physiological circulating level, and the effects caused by this dose range may represent the physiological functions of GnRH [73,74]. At the low concentrations (0.1–10nM) GnRH stimulates cell proliferation, migration and invasion in a dose-dependent manner whereas high concentrations (100nM to 1µM) inhibit these functions [7577]. Moreover, the same dose of GnRH can elicit completely opposite responses in cells derived from the same tissue. In two human ovarian cancer cell lines OVCAR-3 and SKOV-3, GnRH-I and GnRH-II induce invasion of OVCAR-3 cells, but inhibit the invasiveness of the SKOV-3 cells [76]. Similar differences have been found in the effects of GnRH on cell proliferation and cell migration in the prostate carcinoma cell lines TSU-Pr1 and DU-145 [78]. The observation that GnRH-I and GnRH-II have no significant effects on cell lines with type I GnRHR depletion, indicates that the type I GnRHR is indispensable for the effects of both GnRH-I and GnRH-II [76, 78].

Functional differences of GnRHR output may, in-part, be due to the fact that the receptor interacts with different G-proteins and other signaling factors in a ‘cell context’-dependent manner, potentially a generic facet of GPCR signal transduction activity [79]. As we have described, GnRH actions have been shown to be mediated by coupling to different Gα-proteins, depending on the mode of ligand exposure [70, 78]. In general, it has been noted that Gαq and Gαs are associated with GnRH stimulatory effects [72], whereas Gαi often mediates the antiproliferative and proapoptotic effects of GnRH [8183]. Low GnRH concentrations appear to primarily promote the coupling of GnRHR to Gαs [72]. It has also been demonstrated that peptidergic GnRH analogs can exert direct anti-proliferative actions upon reproductive tissues, in which Gαi stimulation has been implicated [8487]. In addition to peptide GnRH analogs and GnRH-I, the role of GnRH-II as an autocrine growth inhibitor has also been demonstrated. Like GnRH-I, treatment with GnRH-II in vitro inhibits the proliferation of both non-tumorigenic and tumorigenic ovarian surface epithelial cells in a dose- and time-dependent manner [12, 88]. These findings suggest that the intracellular milieu in different tissues can result in differential coupling and diverse phenotypic signaling effects, potentially through the creation of unique substates of the Type I GnRH receptor [58, 79].

NEUROPHYSIOLOGICAL ACTIONS OF GnRH

GnRH acts not only within the pituitary gonadotropes, but also exerts receptor-mediated effects in placenta, gonads, immune cells, breast, prostate, as well as neurotransmitter and neuromodulatory actions in the central and peripheral nervous system [12, 8998]. With respect to the pathophysiology of cognitive disorders such as AD, that often originate in the hippocampus, it is interesting to note that the hippocampus is one of the CNS regions with the higher levels of GnRH receptor expression. The hippocampus is one of the most important integrative central nervous regions for cognitive, endocrinological and behavioral processes [99,100]. Hippocampal pyramidal neurons demonstrate immunoreactive GnRH receptors and GnRH has been detected in human hippocampus extractions [101, 102]. Activation of GnRH receptors can induce a long-lasting enhancement of synaptic transmission, mediated by ionotropic glutamate receptors in CA1 pyramidal neurons of rat hippocampal slices. GnRH potentiates the intrinsic neuronal excitability of both hippocampal CA1 and CA3 pyramidal neurons in the hippocampus. GnRH-mediated hippocampal neuron activation has also been shown to be profoundly modified by estrogen levels in the rat [103]. The effects of elevated post-menopausal GnRH, due to the loss of estrogen negative feedback [104], upon hippocampal neurons may constitute a component of the neurodegenerative pathology that accompanies AD [99]. GnRH not only affects the excitability of hippocampal neurons but also has the potential to regulate the excitability of cortical neurons which are also crucially involved in learning and memory. Low levels of GnRH have been reported in the human cortex [105], in addition, the splicing intermediate of mature GnRH mRNA, which still contains intron A, has also been detected in the rat cortex [106,107]. GnRH receptor immunoreactive neurons in the cerebral cortex are widespread, suggesting that GnRH may act as common neuromodulatory peptide for multiple signaling tracts in the CNS [108,109]. In the rat, GnRH depresses the activity of cortical neurons [110,111] and has been shown to affect neurite outgrowth and neurofilament protein expression in cultured cortical neurons [112]. In this regard, it will be interesting to examine the additional structural variants of the GnRH family (GnRH II and GnRH III) with respect to their role in modulating structure and function in the brain. The complete and universal conservation of GnRH IIs amino acid sequence for more than 500 million years suggests that GnRH II may mediate functions vital for most forms of multicellular life. GnRH II appears to have a variety of reproductive and non-reproductive functions. [113,114]. The wide distribution of GnRH II in the central and peripheral nervous systems suggests a neurotransmitter/neuromodulatory role. The first studies of the neuronal actions of GnRH II demonstrated the inhibition of the KCNQ channel (M-current) in the bullfrog sympathetic ganglion which sensitizes neurons to depolarization [115]. In nutritionally compromised musk shrews or female marmosets, GnRH II stimulates reproductive/sexual behavior while interestingly GnRH I is inactive, suggesting that GnRH II is specific for the neurological effects related to behavior [114]. In addition to its role as a generalized neuromodulator in the nervous system, GnRH II is also present in non-neural reproductive tissues, such as the prostate [116]. GnRH binding sites and antiproliferative effects of GnRH analogues have been described in reproductive tissue tumors and their cell lines [58, 117]. Interestingly the GnRH binding sites, signaling and pharmacological effects of GnRH ligands in many peripheral reproductive tissues are distinct from typical pituitary Type I GnRH receptors, but show a greater functional similarity to Type II GnRH receptors from other species [48, 58]. However as we have described, a full-length Type II GnRH receptor is absent from man, chimpanzee, cow, horse, sheep, rat and mouse [48, 118]. The genetic lack of a functional Type II GnRH receptor in specific species is tolerated pharmacologically by the accommodation of GnRH II signaling through a modified ‘allotype’ Type I GnRH receptor, with a distinctly different pharmacological profile [58, 119]. This functional modulation is made by the forced and selective interaction of the receptor in certain tissues with distinct signal transduction systems to those that interact stably with the Type I GnRH receptor in the pituitary. The selective stimulation of this differential type of GnRH receptor can be achieved through the creation of GnRH analogs by extensive chemical alterations of the GnRH I backbone [58, 119].

GnRH AND ALZHEIMER’S DISEASE PATHOPHYSIOLOGY

AD is the most common form of dementia in elderly people. Familial AD is characterized by a marked decline in memory and cognitive performance, including deterioration of language, as well as defects in visual and motor coordination, and eventual death [120, 121]. AD involves a loss of neurons, beginning in the entorhinal cortex and later spreading to the neocortex [122]. AD is not only the predominant cause of senile dementia, but is also the most prevalent neurodegenerative disease worldwide [121]. At the molecular level, familial AD is characterized pathologically by the occurrence of intracellular neurofibrillary tangles rich in tau protein and extracellular plaques containing β-amyloid (Aβ) peptides [123, 124]. These amyloid plaques and neurofibrillary tangles accumulate first in the hippocampal and cortical regions of the brain [125; 126]. In addition, mice transgenically overexpressing Aβ, or containing genetic mutations that enhance Aβ aggregation, show many of the symptoms of AD [127130]. Aβ is the product of serial cleavage of the amyloid precursor protein (APP), first by β and then by γ secretases, to yield Aβ peptides of varying lengths, predominantly the 37-, 40-, and 42- residue forms. An increasing ratio of the full-length, 1–42 peptide to the 1–40 form is associated with disease [124], and mutations underlying familial forms of AD either increase this ratio or increase the amount of Aβ secreted [131]. Aβ peptides belong to a class of natively unfolded proteins, and as a consequence can adopt a wide variety of tertiary and quaternary structures in vivo and in vitro, including monomers, oligomers, and fibrils [132,133]. However, other naturally occurring oligomeric forms of Aβ are also toxic [133,134], and evidence is accumulating that the capacity of Aβ, mutant Aβ, or fragments of Aβ to aggregate into oligomers is directly related to toxicity [131]. In this regard, the leading hypothesis is that amyloid-β deposition causes the disease [135] since familial forms of AD, resulting from mutations in either the amyloid-β protein precursor or presenilins-1/2, all affect the processing of amyloid-β [136]. However, since perturbation of these elements in cell or animal models does not fully result in the multitude of biochemical and cellular changes found in the human disease [137139], it is evident that other factors are also involved. In fact, there is now considerable evidence indicating that amyloid-β may be a consequence rather than causative factor in disease pathogenesis [139141]. Aside from the classical amyloid hypothesis, other theories of AD etiology include: tau phosphorylation; accumulated oxidative stress; mitochondrial alterations; metal ion dysregulation; inflammation. It is clear that these pathologies are involved in the disease process, but the actual upstream cause of these in AD etiology is currently poorly understood. Age-related decline in gonadal reproductive hormones and the integrity of the HPG axis has received considerable attention with regards to their potential role in AD, e.g. women who maintain relatively high endogenous estrogen levels and functional HPG axis after menopause exhibit a decreased prevalence of AD [142]. Findings regarding the benefits of hormone replacement therapy (HRT) in AD however have suggested that falling levels of steroid hormones that accompany menopause/andropause cannot sufficiently explain patterns of AD susceptibility[143]. In fact, it is only when one takes into account the role(s) of other hormones and receptor systems of the HPG axis that the susceptibility, onset, and progression of AD can be accurately characterized, e.g. the GnRH system that can affect hippocampus and cortical function, during the “critical period” around menopause/andropause. Due to the ability of GnRH to directly regulate LH, FSH and sex steroid production/gametogenesis, GnRH plays a central role in both reproductive function and general hormonal control via precise regulation of the HPG axis. It has been demonstrated that GnRH receptor concentration decreases to low levels during adult reproductive life, before paradoxically increasing in older rats (17 and 21 months of age). This increase in GnRH receptor concentration in post-reproductive animals appears to be due to decreased gonadal hormone production, since castrated male and female rats (high LH/FSH, low estrogen/testosterone) display increased GnRH receptor expression. Linked to this, it has been shown that male and female rats treated with testosterone and estradiol/ progesterone have decreased GnRH receptor expression [28, 144]. With regards to the dynamic reproductive status in animals, GnRH receptor concentration is highest in proestrus and significantly lower during estrus [145]. In addition to steroid-induced changes in GnRH receptor expression, the affinity of GnRH receptor for GnRH decreases 18-fold during diestrus I and estrus, compared to ovariectomized animals [145]. In this review, we propose that GnRH, in addition to estrogen, may also be a prime factor in the pathogenesis of AD. Although the concentration of circulating GnRH is very low due to its short halflife [146, 147], there is a two fold increase in circulating gonadotropins in individuals with AD, compared with age-matched control individuals [148, 149]. GnRH therefore appears to possess a strong biochemical and electrophysiological action in hippocampal neurons [102, 103, 144, 150]. These actions seem to be connected to and modulated by the presence of estrogen however [103]. The effect of GnRH, with or without the presence of estrogen, on hippocampal pyramidal neurons may constitute an important component of the neurodegenerative pathology that accompanies AD [104]. It is notable that expression of hippocampal spinophilin, a reliable dendritic spine marker, linked to cognitive status that functionally interacts and scaffolds GPCRs, can be directly regulated by the GnRH system [151]. The hippocampal GnRH system is itself acutely sensitive to both age and reproductive status, as hippocampal GnRH receptor expression is increased in aged and castrated rats [28, 103].

If the pathology of AD is linked to disruption of the HPG axis, another strategy to suppress the increase in the gonadotropins following menopause/andropause is to use GnRH analogues (agonists and antagonists), which can decrease serum gonadotropin levels and also potentially have direct neuroprotective CNS actions. It has been demonstrated that administration of leuprolide acetate to C57BL/6 mice produced significant decreases in brain Aβ [23] and was also shown to improve cognitive function in a transgenic mouse model of AD that correlated to decreases in the Aβ burden [152]. The latter study used mice at a very advanced stage of cognitive decline, indicating a profound therapeutic efficacy of GnRH-based therapies, even in advanced stages of AD pathophysiology. Therefore, despite often complex age-dependent alterations in the GnRH system, targeting this receptor-ligand system may well be an effective strategy to combat the incidence as well as the progression of AD.

CONCLUSION

Following early reproductive life, changes in serum and neuronal concentrations of HPG axis hormones (increased gonadotropins, increased activins, increased GnRH and decreased sex steroids) associated with menopause/andropause have the potential to profoundly alter qualitatively and quantitatively neuronal signaling mechanisms. In particular, alterations in the GnRH-regulated LH release promotes biochemical and cellular changes consistent with the neurodegenerative changes observed in the AD brain [153]. These findings support the premise that GnRH receptor-based therapeutics could be a potential therapeutic target for the treatment of AD. Several double-blind placebo controlled phase II clinical trials are currently underway to conclusively make this determination. A reconsideration of the potential widespread actions that this traditional reproductive hormone exerts may lead to the generation of novel therapies and provide insight into the dynamic temporal alterations of GnRH signaling in normal and pathological aging. To ensure tissue specificity, the rational design of new therapeutics should take into account recent advances in our understanding of GnRH and generic receptor signaling mechanisms [58, 79, 154157]. Taken together, the multiple findings of GnRH functionality in cognition may facilitate the future creation of novel selective GnRH analogs with potential for wider and more specific application to multiple neurodegenerative diseases.

ACKNOWLEDGEMENTS

This research was supported by the Intramural Research Program of the NIH, National Institute on Aging. The authors have no conflicts of scientific interest with respect to the manuscript.

ABBREVIATIONS

HPG

The hypothalamic-pituitary-gonadal

AD

Alzheimer’s disease

GnRH

Gonadotropin-releasing hormone

GPCR

G protein-coupled receptor

Aβ

beta-amyloid peptide

cAMP

cyclic adenosine monophosphate

DAG

diacylglycerol

ERK

extracellular signal-regulated kinase

FSH

follicle stimulating hormone

G-protein

guanine nucleotide binding protein

GnRH

gonadotropin-releasing hormone

GnRHR

gonadotropin- releasing hormone receptor

GTP

guanosine triphosphate

G-protein α subunit

Gβγ

G-protein βγ subunits

IP3

inositol 1, 4, 5-trisphosphate

JNK

jun-N-terminal kinase

LH

luteinizing hormone

LPA

lysophosphatidic acid

MAPK

mitogen-activated protein kinase

PIP2

phosphatidylinositol l-4-5-bisphosphate

PL

phospholipase

PLA2

phospholipase A2

PLCβ

phospholipase Cβ

PLD

phospholipase D

PKC

protein kinase C

RGS

regulator of G-protein signaling

HRT

hormone replacement therapy

APP

the amyloid precursor protein

MAPK

mitogen-activated protein kinase

REFFERENCES

  • 1.Millar RP, Lu ZL, Pawson AJ, Flanagan CA, Morgan K, Maudsley SR. Gonadotropin-releasing hormone receptors. Endocr. Rev. 2004;25:235–275. doi: 10.1210/er.2003-0002. [DOI] [PubMed] [Google Scholar]
  • 2.Caraty A, Locatelli A. Effect of time after castration on secretion of LHRH and LH in the ram. J. Reprod. Fertil. 1988;82:263–269. doi: 10.1530/jrf.0.0820263. [DOI] [PubMed] [Google Scholar]
  • 3.Jan YN, Jan LY, Kuffler SW. A peptide as a possible transmitter in sympathetic ganglia of the frog. Proc. Natl. Acad. Sci. USA. 1979;76:1501–1505. doi: 10.1073/pnas.76.3.1501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Cheung LW, Wong AS. Gonadotropin-releasing hormone: GnRH receptor signaling in extrapituitary tissues. FEBS J. 2008;275:5479–5495. doi: 10.1111/j.1742-4658.2008.06677.x. [DOI] [PubMed] [Google Scholar]
  • 5.Jan YN, Jan LY, Kuffler SW. Further evidence for peptidergic transmission in sympathetic ganglia. Proc Natl. Acad. Sci. USA. 1980;77:5008–5012. doi: 10.1073/pnas.77.8.5008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Yang SN, Lu F, Wu JN, Liu DD, Hsieh WY. Activation of gonadotropin-releasing hormone receptors induces a long-term enhancement of excitatory postsynaptic currents mediated by ionotropic glutamate receptors in the rat hippocampus. Neurosci. Lett. 1999;260:33–36. doi: 10.1016/s0304-3940(98)00939-2. [DOI] [PubMed] [Google Scholar]
  • 7.Almeida OP, Waterreus A, Spry N, Flicker L, Martins RN. One year follow-up study of the association between chemical castration, sex hormones, beta-amyloid, memory and depression in men. Psychoneuroendocrinology. 2004;29(8):1071–1081. doi: 10.1016/j.psyneuen.2003.11.002. [DOI] [PubMed] [Google Scholar]
  • 8.Gandy S, Almeida OP, Fonte J, Lim D, Waterrus A, Spry N, Flicker L, Martins RN. Chemical andropause and amyloid-beta peptide. JAMA. 2001;285:2195–2196. doi: 10.1001/jama.285.17.2195-a. [DOI] [PubMed] [Google Scholar]
  • 9.Troen BR. The biology of aging. Mt. Sinai J. Med. 2003;70(1):3–22. [PubMed] [Google Scholar]
  • 10.Boulianne GL. Neuronal regulation of lifespan: clues from flies and worms. Mech. Ageing Dev. 2001;122:883–894. doi: 10.1016/s0047-6374(01)00245-7. [DOI] [PubMed] [Google Scholar]
  • 11.Martin B, Golden E, Carlson OD, Egan JM, Mattson MP, Maudsley S. Caloric restriction: impact upon pituitary function and reproduction. Ageing Res. Rev. 2008;7:209–224. doi: 10.1016/j.arr.2008.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Millar RP, King JA, Davidson JS, Milton RC. Gonadotrophin-releasing hormone--diversity of functions and clinical applications. S. Afr. Med. J. 1987;72(11):748–755. [PubMed] [Google Scholar]
  • 13.Hsueh AJ, Schaeffer JM. Gonadotropin-releasing hormone as a paracrine hormone and neurotransmitter in extra-pituitary sites. J. Steroid. Biochem. 1985;23:757–764. doi: 10.1016/s0022-4731(85)80011-x. [DOI] [PubMed] [Google Scholar]
  • 14.Jennes L, Conn PM. Gonadotropin-releasing hormone and its receptors in rat brain. Front. Neuroendocrinol. 1994;15:51–77. doi: 10.1006/frne.1994.1003. [DOI] [PubMed] [Google Scholar]
  • 15.Millar RP, Lu ZL, Pawson AJ, Flanagan CA, Morgan K, Maudsley SR. Gonadotropin-releasing hormone receptors. Endocr. Rev. 2004;25:235–275. doi: 10.1210/er.2003-0002. [DOI] [PubMed] [Google Scholar]
  • 16.Couzinet B, Schaison G. The control of gonadotrophin secretion by ovarian steroids. Hum. Reprod. 1993;8:97–101. doi: 10.1093/humrep/8.suppl_2.97. [DOI] [PubMed] [Google Scholar]
  • 17.Chakravarti S, Collins WP, Forecast JD, Newton JR, Oram DH, Studd JW. Hormonal profiles after the menopause. Br. Med. J. 1976;2:784–787. doi: 10.1136/bmj.2.6039.784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Wide L, Nillius SJ, Gemzell C, Roos P. Radioimmunosorbent assay of follicle-stimulating hormone and luteinizing hormone in serum and urine from men and women. Acta. Endocrinol. Suppl. 1973;174:3–58. [PubMed] [Google Scholar]
  • 19.Neaves WB, Johnson L, Porter JC, Parker CR, Jr, Petty CS. Leydig cell numbers, daily sperm production, and serum gonadotropin levels in aging men. J. Clin. Endocrinol. Metab. 1984;59:756–763. doi: 10.1210/jcem-59-4-756. [DOI] [PubMed] [Google Scholar]
  • 20.Nunomura A, Perry G, Aliev G, Hirai K, Takeda A, Balraj EK, Jones PK, Ghanbari H, Wataya T, Shimohama S, Chiba S, Atwood CS, Petersen RB, Smith MA. Oxidative damage is the earliest event in Alzheimer disease. J. Neuropathol. Exp. Neurol. 2001;60:759–767. doi: 10.1093/jnen/60.8.759. [DOI] [PubMed] [Google Scholar]
  • 21.Ogawa O, Lee HG, Zhu X, Raina A, Harris PL, Castellani RJ, Perry G, Smith MA. Increased p27, an essential component of cell cycle control, in Alzheimer's disease. Aging Cell. 2003;2:105–110. doi: 10.1046/j.1474-9728.2003.00042.x. [DOI] [PubMed] [Google Scholar]
  • 22.Harris D, Bonfil D, Chuderland D, Kraus S, Seger R, Naor Z. Activation of MAPK cascades by GnRH: ERK and Jun N-terminal kinase are involved in basal and GnRH-stimulated activity of the glycoprotein hormone LHbeta-subunit promoter. Endocrinology. 2002;143:1018–1025. doi: 10.1210/endo.143.3.8675. [DOI] [PubMed] [Google Scholar]
  • 23.Bowen RL, Smith MA, Harris PL, Kubat Z, Martins RN, Castellani RJ, Perry G, Atwood CS. Elevated luteinizing hormone expression colocalizes with neurons vulnerable to Alzheimer's disease pathology. J. Neurosci. Res. 2002;70:514–518. doi: 10.1002/jnr.10452. [DOI] [PubMed] [Google Scholar]
  • 24.Mattson MP, Maudsley S, Martin B. A neural signaling triumvirate that influences ageing and age-related disease: insulin/IGF-1, BDNF and serotonin. Ageing Res Rev. 2004;3(4):445–464. doi: 10.1016/j.arr.2004.08.001. [DOI] [PubMed] [Google Scholar]
  • 25.Powell JF, Reska-Skinner SM, Prakash MO, Fischer WH, Park M, Rivier JE, Craig AG, Mackie GO, Sherwood NM. Two new forms of gonadotropin-releasing hormone in a protochordate and the evolutionary implications. Proc. Natl. Acad. Sci. U.S.A. 1996;93:10461–10464. doi: 10.1073/pnas.93.19.10461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Adams BA, Tello JA, Erchegyi J, Warby C, Hong DJ, Akinsanya KO, Mackie GO, Vale W, Rivier JE, Sherwood NM. Six novel gonadotropin-releasing hormones are encoded as triplets on each of two genes in the protochordate, Ciona intestinalis. Endocrinology. 2003;144:1907–1919. doi: 10.1210/en.2002-0216. [DOI] [PubMed] [Google Scholar]
  • 27.Badr M, Marchetti B, Pelletier G. Changes in hippocampal LH-RH receptor density during maturation and aging in the rat. Brain Res. Dev. Brain Res. 1989;45(2):179–184. doi: 10.1016/0165-3806(89)90037-0. [DOI] [PubMed] [Google Scholar]
  • 28.Badr M, Marchetti B, Pelletier G. Modulation of hippocampal LHRH receptors by sex steroids in the rat. Peptides. 1988;9(2):441–442. doi: 10.1016/0196-9781(88)90283-5. [DOI] [PubMed] [Google Scholar]
  • 29.Ehret G, Buckenmaier J. Estrogen-receptor occurrence in the female mouse brain: effects of maternal experience, ovariectomy, estrogen and anosmia. J Physiol Paris. 1994;88(5):315–329. doi: 10.1016/0928-4257(94)90012-4. [DOI] [PubMed] [Google Scholar]
  • 30.Adams MM, Fink SE, Shah RA, Janssen WG, Hayashi S, Milner TA, McEwen BS, Morrison JH. Estrogen and aging affect the subcellular distribution of estrogen receptor-alpha in the hippocampus of female rats. J Neurosci. 2002;22(9):3608–3614. doi: 10.1523/JNEUROSCI.22-09-03608.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Sar M, Lubahn DB, French FS, Wilson EM. Immunohistochemical localization of the androgen receptor in rat and human tissues. Endocrinology. 1990;127(6):3180–3186. doi: 10.1210/endo-127-6-3180. [DOI] [PubMed] [Google Scholar]
  • 32.Xiao L, Jordan CL. Sex differences, laterality, and hormonal regulation of androgen receptor immunoreactivity in rat hippocampus. Horm Behav. 2002;42(3):327–336. doi: 10.1006/hbeh.2002.1822. [DOI] [PubMed] [Google Scholar]
  • 33.Henderson VW, Paganini-Hill A, Emanuel CK, Dunn ME, Buckwalter JG. Estrogen Replacement Therapy in Older Women: Comparisons Between Alzheimer's Disease Cases and Nondemented Control Subjects. Arch. Neurol. 1994;51:896–900. doi: 10.1001/archneur.1994.00540210068014. [DOI] [PubMed] [Google Scholar]
  • 34.Tang MX, Jacobs D, Stern Y, Marder K, Schofield P, Gurland B, Andrews H, Mayeux R. Effect of oestrogen during menopause on risk and age at onset of Alzheimer's disease. Lancet. 1996;348:429–432. doi: 10.1016/S0140-6736(96)03356-9. [DOI] [PubMed] [Google Scholar]
  • 35.Fratiglioni L, Viitanen M, von Strauss E, Tontodonati V, Herlitz A, Winblad B. Very Old Women at Highest Risk of Dementia and Alzheimer's Disease: Incidence Data from the Kungsholmen Project, Stockholm. Neurology. 1997;48:132–138. doi: 10.1212/wnl.48.1.132. [DOI] [PubMed] [Google Scholar]
  • 36.Stam FC, Wigboldus JM, Smeulders AW. Age incidence of senile brain amyloidosis. Pathol. Res. Pract. 1986;181(5):558–562. doi: 10.1016/S0344-0338(86)80149-2. [DOI] [PubMed] [Google Scholar]
  • 37.Jorm AF, Korten AE, Henderson AS. The prevalence of dementia: A quantitative integration of the literature. Acta Psychiatr Scand. 1987;76:465–479. doi: 10.1111/j.1600-0447.1987.tb02906.x. [DOI] [PubMed] [Google Scholar]
  • 38.Writing Group for the Women’s Health Initiative Investigators. Risks and benefits of estrogen plus progestin in healthy postmenopausal women. JAMA. 2002;288:321–333. doi: 10.1001/jama.288.3.321. [DOI] [PubMed] [Google Scholar]
  • 39.Bless EP, Walker HJ, Yu KW, Knoll JG, Moenter SM, Schwarting GA, et al. Live view of gonadotropin - releasing hormone containing neuron migration. Endocrinology. 2005;146:463–468. doi: 10.1210/en.2004-0838. [DOI] [PubMed] [Google Scholar]
  • 40.Song T, Nikolics K, Seeburg PH, Goldsmith PC. GnRH-prohormone-containing neurons in the primate brain: immunostaining for the GnRH-associated peptide. Peptides. 1987;8:335–346. doi: 10.1016/0196-9781(87)90109-4. [DOI] [PubMed] [Google Scholar]
  • 41.Kah O, Lethimonier C, Somoza G, Guilgur LG, Vaillant C, Lareyre JJ. GnRH and GnRH receptors in metazoa: A historical, comparative, and evolutive perspective. Gen. Comp. Endocrinol. 2007;153:346–364. doi: 10.1016/j.ygcen.2007.01.030. [DOI] [PubMed] [Google Scholar]
  • 42.Silver MR, Kawauchi H, Nozaki M, Sower SA. Cloning and analysis of the lamprey GnRH-III cDNA from eight species of lamprey representing the three families of Petromyzoniformes. Gen. Comp. Endocrinol. 2004;139:85–94. doi: 10.1016/j.ygcen.2004.07.011. [DOI] [PubMed] [Google Scholar]
  • 43.Suzuki K, Gamble RL, Sower SA. Multiple transcripts encoding lamprey gonadotropin-releasing hormone-I precursore. J. Mol. Endocrinol. 2000;24:365–376. doi: 10.1677/jme.0.0240365. [DOI] [PubMed] [Google Scholar]
  • 44.Amemiya Y, Sogabe Y, Nozaki M, Takahashi A, Kawauchi H. Somatolactin in the White Sturgeon and African Lungfish and Its Evolutionary Significance. Gen. Comp. Endocrinol. 1999;114:181–190. doi: 10.1006/gcen.1998.7250. [DOI] [PubMed] [Google Scholar]
  • 45.Gorbman A, Sower SA. Evolution of the role of GnRH in animal (Metazoan) biology. Gen. Comp. Endocrinol. 2003;134:207–213. doi: 10.1016/j.ygcen.2003.09.018. [DOI] [PubMed] [Google Scholar]
  • 46.Lescheid DW, Terasawa E, Abler LA, Urbanski HF, Warby CM, Millar RP, Sherwood NM. A Second Form of Gonadotropin-Releasing Hormone (GnRH) with Characteristics of Chicken GnRH-II Is Present in the Primate Brain. Endocrinology. 1997;138:5618–5629. doi: 10.1210/endo.138.12.5592. [DOI] [PubMed] [Google Scholar]
  • 47.Trinh LA, McCutchen MD, Bonner-Fraser M, Fraser SE, Bumm LA, McCauley DW. Fluorescent in situ hybridization employing the conventional NBT/BCIP chromogenic stain. Biotechniques. 2007;42:756–759. doi: 10.2144/000112476. [DOI] [PubMed] [Google Scholar]
  • 48.Millar RP, Lowe S, Conklin D, Pawson A, Maudsley S, Troskie B, Ott T, Millar M, Lincoln G, Sellar R, Faurholm B, Scobie G, Kuestner R, Terasawa E, Katz A. A novel mammalian receptor for the evolutionarily conserved type II gonadotropin-releasing hormone. Proc. Natl. Acad. Sci. USA. 2001;98:9636–9641. doi: 10.1073/pnas.141048498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Gault PM, Maudsley S, Lincoln GA. Evidence that gonadotropin-releasing hormone II is not a physiological regulator of gonadotropin secretion in mammals. J. Neuroendocrinol. 2003;15:831–839. doi: 10.1046/j.1365-2826.2003.01065.x. [DOI] [PubMed] [Google Scholar]
  • 50.Pawson AJ, Faccenda E, Maudsley S, Lu ZL, Naor Z, Millar RP. Mammalian type I gonadotropin-releasing hormone receptors undergo slow, constitutive, agonist-independent internalization. Endocrinology. 2008;149:1415–1422. doi: 10.1210/en.2007-1159. [DOI] [PubMed] [Google Scholar]
  • 51.Naor Z. Signaling by G-protein-coupled receptor (GPCR): Studies on the GnRH receptor. Front. Neuroendocrinol. 2009;30:10–29. doi: 10.1016/j.yfrne.2008.07.001. [DOI] [PubMed] [Google Scholar]
  • 52.Mitchell R, Sim PJ, Leslie T, Johnson MS, Thomson FJ. Activation of MAP kinase associated with the priming effect of LHRH. J. Endocrinol. 1994;140:8–9. doi: 10.1677/joe.0.140r015. [DOI] [PubMed] [Google Scholar]
  • 53.Reiss N, Llevi LN, Shacham S, Harris D, Seger R, Naor Z. Mechanism of mitogen-activated protein kinase activation by gonadotropin-releasing hormone in the pituitary of alphaT3–1 cell line: differential roles of calcium and protein kinase C. Endocrinology. 1997;138:1673–1682. doi: 10.1210/endo.138.4.5057. [DOI] [PubMed] [Google Scholar]
  • 54.Burger LL, Haisenleder DJ, Dalkin AC, Marshall JC. Regulation of gonadotropin subunit gene transcription. J. Mol. Endocrinol. 2004;33:559–584. doi: 10.1677/jme.1.01600. [DOI] [PubMed] [Google Scholar]
  • 55.Haisenleder DJ, Yasin M, Dalkin AC, Gilrain C, Marshall JC. GnRH regulates steroidogenic factor-1 (SF-1) gene expression in the rat pituitary. Endocrinology. 1996;137:5719–5722. doi: 10.1210/endo.137.12.8940405. [DOI] [PubMed] [Google Scholar]
  • 56.Kaiser UB, Halvorson LM, Chen MT. Sp1, steroidogenic factor 1 (SF-1), and early growth response protein 1 (egr-1) binding sites form a tripartite gonadotropin-releasing hormone response element in the rat luteinizing hormone-beta gene promoter: an integral role for SF-1. Mol Endocrinol. 2000;14(8):1235–1245. doi: 10.1210/mend.14.8.0507. [DOI] [PubMed] [Google Scholar]
  • 57.Borgeat P, Chavancy G, Dupont A, Labrie F, Arimura A, Schally AV. Stimulation of adenosine 30,50-cyclic monophosphate accumulation in anterior pituitary gland in vitro by synthetic luteinizing hormone-releasing hormone. Proc. Natl. Acad. Sci. USA. 1972;69:2677–2681. doi: 10.1073/pnas.69.9.2677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Maudsley S, Davidson L, Pawson AJ, Chan R, López de Maturana R, Millar RP. Gonadotropin-releasing hormone (GnRH) antagonists promote proapoptotic signaling in peripheral reproductive tumor cells by activating a Galphai-coupling state of the type I GnRH receptor. Cancer Res. 2004;64:7533–7544. doi: 10.1158/0008-5472.CAN-04-1360. [DOI] [PubMed] [Google Scholar]
  • 59.Gardner S, Maudsley S, Millar RP, Pawson AJ. Nuclear stabilization of beta-catenin and inactivation of glycogen synthase kinase-3beta by gonadotropin-releasing hormone: targeting Wnt signaling in the pituitary gonadotrope. Mol. Endocrinol. 2007;21:3028–3038. doi: 10.1210/me.2007-0268. [DOI] [PubMed] [Google Scholar]
  • 60.Davidson L, Pawson AJ, Millar RP, Maudsley S. Cytoskeletal reorganization dependence of signaling by the gonadotropin-releasing hormone receptor. J. Biol. Chem. 2004;279:1980–1993. doi: 10.1074/jbc.M309827200. [DOI] [PubMed] [Google Scholar]
  • 61.Maudsley S, Naor Z, Bonfil D, Davidson L, Karali D, Pawson AJ, Larder R, Pope C, Nelson N, Millar RP, Brown P. Proline-rich tyrosine kinase 2 mediates gonadotropin-releasing hormone signaling to a specific extracellularly regulated kinase-sensitive transcriptional locus in the luteinizing hormone beta-subunit gene. Mol. Endocrinol. 2007;21:1216–1233. doi: 10.1210/me.2006-0053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Davidson L, Pawson AJ, López de Maturana R, Freestone SH, Barran P, Millar RP, Maudsley S. Gonadotropin-releasing hormone-induced activation of diacylglycerol kinase-zeta and its association with active c-Src. J. Biol. Chem. 2004;279:11906–11916. doi: 10.1074/jbc.M310784200. [DOI] [PubMed] [Google Scholar]
  • 63.Troskie B, King JA, Millar RP, Peng YY, Kim J, Figueras H, Illing N. Chicken GnRH II-like peptides and a GnRH receptor selective for chicken GnRH II in amphibian sympathetic ganglia. Neuroendocrinology. 1997;65:396–402. doi: 10.1159/000127202. [DOI] [PubMed] [Google Scholar]
  • 64.Pawson AJ, Maudsley S, Morgan K, Davidson L, Naor Z, Millar RP. Inhibition of human type I gonadotropin-releasing hormone receptor (GnRHR) function by expression of a human type II GnRHR gene fragment. Endocrinology. 2005;146:2639–2649. doi: 10.1210/en.2005-0133. [DOI] [PubMed] [Google Scholar]
  • 65.Berman DM, Wilkie TM, Gilman AG. GAIP and RGS4 are GTPase-activating proteins for the Gi subfamily of G protein alpha subunits. Cell. 1996;86:445–452. doi: 10.1016/s0092-8674(00)80117-8. [DOI] [PubMed] [Google Scholar]
  • 66.Maudsley S, Davidson L, Pawson AJ, Freestone SH, López de Maturana R, Thomson AA, Millar RP. Gonadotropin-releasing hormone functionally antagonizes testosterone activation of the human androgen receptor in prostate cells through focal adhesion complexes involving Hic-5. Neuroendocrinology. 2006;84(5):285–300. doi: 10.1159/000098402. [DOI] [PubMed] [Google Scholar]
  • 67.Maudsley S, Pierce KL, Zamah AM, Miller WE, Ahn S, Daaka Y, Lefkowitz RJ, Luttrell LM. The beta(2)-adrenergic receptor mediates extracellular signal-regulated kinase activation via assembly of a multi-receptor complex with the epidermal growth factor receptor. J. Biol. Chem. 2000;275:9572–9580. doi: 10.1074/jbc.275.13.9572. [DOI] [PubMed] [Google Scholar]
  • 68.Shah BH, Soh JW, Catt KJ. Dependence of gonadotropin-releasing hormone-induced neuronal MAPK signaling on epidermal growth factor receptor transactivation. J. Biol. Chem. 2003;278:2866–2875. doi: 10.1074/jbc.M208783200. [DOI] [PubMed] [Google Scholar]
  • 69.Shah BH, Farshori MP, Jambusaria A, Catt KJ. Roles of Src and epidermal growth factor receptor transactivation in transient and sustained ERK1/2 responses to gonadotropin-releasing hormone receptor activation. J. Biol. Chem. 2003;278:19118–19126. doi: 10.1074/jbc.M212932200. [DOI] [PubMed] [Google Scholar]
  • 70.Kraus S, Benard O, Naor Z, Seger R. c-Src is activated by the epidermal growth factor receptor in a pathway that mediates JNK and ERK activation by gonadotropin-releasing hormone in COS7 cells. J. Biol. Chem. 2003;278:32618–32630. doi: 10.1074/jbc.M303886200. [DOI] [PubMed] [Google Scholar]
  • 71.Haisenleder DJ, Cox ME, Parsons SJ, Marshall JC. Gonadotropin-Releasing Hormone Pulses Are Required to Maintain Activation of Mitogen-Activated Protein Kinase: Role in Stimulation of Gonadotrope Gene Expression. Endocrinology. 1998;139:3104–3111. doi: 10.1210/endo.139.7.6091. [DOI] [PubMed] [Google Scholar]
  • 72.Krsmanovic LZ, Mores N, Navarro CE, Arora KK, Catt KJ. An agonist-induced switch in G protein coupling of the gonadotropin-releasing hormone receptor regulates pulsatile neuropeptide secretion. Proc. Natl. Acad. Sci. USA. 2003;100:2969–2974. doi: 10.1073/pnas.0535708100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Enomoto M, Seong JY, Kawashima S, Park MK. Proliferation of TSU-Pr1, a human prostatic carcinoma cell line is stimulated by gonadotropin-releasing hormone. Life Sci. 2004;74:3141–3152. doi: 10.1016/j.lfs.2003.11.009. [DOI] [PubMed] [Google Scholar]
  • 74.Loumaye E, Catt KJ. Homologous regulation of gonadotropin-releasing hormone receptors in cultured pituitary cells. Science. 1982;215:983–985. doi: 10.1126/science.6296998. [DOI] [PubMed] [Google Scholar]
  • 75.Cheung LW, Leung PC, Wong AS. Gonadotropin-Releasing Hormone Promotes Ovarian Cancer Cell Invasiveness through c-Jun NH2-Terminal Kinase–Mediated Activation of Matrix Metalloproteinase (MMP)-2 and MMP-9. Cancer Res. 2006;66:10902–10910. doi: 10.1158/0008-5472.CAN-06-2217. [DOI] [PubMed] [Google Scholar]
  • 76.Chen CL, Cheung LW, Lau MT, Choi JH, Auersperg N, Wang HS, Wong AS, Leung PC. Differential role of gonadotropin-releasing hormone on human ovarian epithelial cancer cell invasion. Endocrine. 2007;31:311–320. doi: 10.1007/s12020-007-0041-8. [DOI] [PubMed] [Google Scholar]
  • 77.Arencibia JM, Schally AV. Luteinizing hormone-releasing hormone as an autocrine growth factor in ES-2 ovarian cancer cell line. Int. J. Oncol. 2000;16:1009–1013. doi: 10.3892/ijo.16.5.1009. [DOI] [PubMed] [Google Scholar]
  • 78.Enomoto M, Utsumi M, Park MK. Gonadotropin-Releasing Hormone Induces Actin Cytoskeleton Remodeling and Affects Cell Migration in a Cell-Type-Specific Manner in TSU-Pr1 and DU145 Cells. Endocrinology. 2006;147:530–542. doi: 10.1210/en.2005-0460. [DOI] [PubMed] [Google Scholar]
  • 79.Maudsley S, Martin B, Luttrell LM. The origins of diversity and specificity in G protein-coupled receptor signaling. J. Pharmacol. Exp. Ther. 2005;314:485–494. doi: 10.1124/jpet.105.083121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Liu F, Usui I, Evans LG, Austin DA, Mellon PL, Olefsky JM, Webster NJ. Involvement of both G(q / 11) and G(s) proteins in gonadotropin-releasing hormone receptor-mediated signaling in L beta T2 cells. J. Biol. Chem. 2002;277:32099–32108. doi: 10.1074/jbc.M203639200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Kraus S, Naor Z, Seger R. Gonadotropin-releasing hormone in apoptosis of prostate cancer cells. Cancer Lett. 2006;234:109–123. doi: 10.1016/j.canlet.2005.02.038. [DOI] [PubMed] [Google Scholar]
  • 82.Imai A, Horibe S, Takagi A, Tamaya T. Gi protein activation of gonadotropin-releasing hormone–mediated protein dephosphorylation in human endometrial carcinoma. Am. J. Obstet. Gynecol. 1997;176:371–376. doi: 10.1016/s0002-9378(97)70501-5. [DOI] [PubMed] [Google Scholar]
  • 83.Raga F, Casan EM, Wen Y, Huang HY, Bonilla- Musoles F, Polan ML. Independent Regulation of Matrix Metalloproteinase-9, Tissue Inhibitor of Metalloproteinase-1 (TIMP-1), and TIMP-3 in Human Endometrial Stromal Cells by Gonadotropin-Releasing Hormone: Implications in Early Human Implantation. J. Clin. Endocrinol. Metab. 1999;84:636–642. doi: 10.1210/jcem.84.2.5464. [DOI] [PubMed] [Google Scholar]
  • 84.Chou CS, Zhu H, Shalev E, MacCalman CD, Leung PC. The effects of gonadotrophin-releasing hormone (GnRH) I and GnRH II on the urokinase-type plasminogen activator inhibitor system in human extravillous cytotrophoblasts in vitro. J. Clin. Endocrinol. Metab. 2002;87:5594–5603. doi: 10.1210/jc.2002-020883. [DOI] [PubMed] [Google Scholar]
  • 85.Moretti RM, Montagnani Marelli M, Van Groeninghen JC, Limonta P. The Effects of Gonadotropin-Releasing Hormone (GnRH) I and GnRH II on the Urokinase-Type Plasminogen Activator/Plasminogen Activator Inhibitor System in Human Extravillous Cytotrophoblasts in Vitro. J. Clin. Endocrinol. Metab. 2002;87:3791–3797. [Google Scholar]
  • 86.Moretti RM, Monagnani Marelli M, van Groeninghen JC, Motta M, Limonta P. Inhibitory activity of luteinizing hormone-releasing hormone on tumor growth and progression. Endocr. Relat. Cancer. 2003;10:161–167. doi: 10.1677/erc.0.0100161. [DOI] [PubMed] [Google Scholar]
  • 87.Choi KC, Auersperg N, Leung PC. Expression and antiproliferative effect of a second form of gonadotropin-releasing hormone in normal and neoplastic ovarian surface epithelial cells. J. Clin. Endocrinol. Metab. 2001;86(10):5075–5078. doi: 10.1210/jcem.86.10.8100. [DOI] [PubMed] [Google Scholar]
  • 88.Kim KY, Choi KC, Park SH, Auersperg N, Leung PC. Extracellular signal-regulated protein kinase, but not c-Jun N-terminal kinase, is activated by type II gonadotrophin-releasing hormone involved in the inhibition of ovarian cancer cell proliferation. J. Clin. Endocrinol. Metab. 2005;90:1670–1677. doi: 10.1210/jc.2004-1636. [DOI] [PubMed] [Google Scholar]
  • 89.Rissman EF, Li X, King JA, Millar RP. Behavioral regulation of gonadotropin-releasing hormone production. Brain Res. Bull. 1997;44(4):459–464. doi: 10.1016/s0361-9230(97)00226-8. [DOI] [PubMed] [Google Scholar]
  • 90.Emons G, Schally AV. The use of luteinizing hormone releasing hormone agonists and antagonists in gynaecological cancers. Hum. Reprod. 1994;9(7):1364–1379. doi: 10.1093/oxfordjournals.humrep.a138714. [DOI] [PubMed] [Google Scholar]
  • 91.King JA, Millar RP, Vallarino M, Pierantoni R. Localization and characterization of gonadotropin-releasing hormones in the brain, gonads, and plasma of a dipnoi (lungfish, Protopterus annectens) Regul. Pept. 1995;57(2):163–174. doi: 10.1016/0167-0115(95)00025-7. [DOI] [PubMed] [Google Scholar]
  • 92.Troskie B, King JA, Millar RP, Peng YY, Kim J, Figueras H, Illing N. Chicken GnRH II-like peptides and a GnRH receptor selective for chicken GnRH II in amphibian sympathetic ganglia. Neuroendocrinology. 1997;65(6):396–402. doi: 10.1159/000127202. [DOI] [PubMed] [Google Scholar]
  • 93.Sower SA, King JA, Millar RP, Sherwood NM, Marshak DR. Comparative biological properties of lamprey gonadotropin-releasing hormone in vertebrates. Endocrinology. 1987;120(2):773–779. doi: 10.1210/endo-120-2-773. [DOI] [PubMed] [Google Scholar]
  • 94.Lovejoy DA, Sherwood NM. Gonadotropin-releasing hormone in ratfish (Hydrolagus colliei): distribution between the sexes and possible relationship with chicken II and salmon II forms. Comp. Biochem. Physiol. B. 1989;92(1):111–118. doi: 10.1016/0305-0491(89)90321-0. [DOI] [PubMed] [Google Scholar]
  • 95.Hsueh AJ, Schaeffer JM. Gonadotropin-releasing hormone as a paracrine hormone and neurotransmitter in extra-pituitary sites. J. Steroid. Biochem. 1985;23(5B):757–764. doi: 10.1016/s0022-4731(85)80011-x. [DOI] [PubMed] [Google Scholar]
  • 96.Jennes L, Conn PM. Gonadotropin-releasing hormone and its receptors in rat brain. Front. Neuroendocrinol. 1994;15(1):51–77. doi: 10.1006/frne.1994.1003. [DOI] [PubMed] [Google Scholar]
  • 97.Sealfon SC, Weinstein H, Millar RP. Molecular mechanisms of ligand interaction with the gonadotropin-releasing hormone receptor. Endocr. Rev. 1997;18:180–207. doi: 10.1210/edrv.18.2.0295. [DOI] [PubMed] [Google Scholar]
  • 98.Wilson AC, Salamat MS, Haasl RJ, Roche KM, Karande A, Meethal SV, Terasawa E, Bowen RL, Atwood CS. Human neurons express type I GnRH receptor and respond to GnRH I by increasing luteinizing hormone expression. J. Endocrinol. 2006;191:651–663. doi: 10.1677/joe.1.07047. [DOI] [PubMed] [Google Scholar]
  • 99.Kubek M, Wilber JF, Leesthma J. The identification of gonadotropin-releasing hormone (GnRH) in hypothalamic and extrahypothalamic loci of the human nervous system. Horm Metab Res. 1979;11:26–29. doi: 10.1055/s-0028-1092676. [DOI] [PubMed] [Google Scholar]
  • 100.He D, Funabashi T, Sano A, Uemura T, Minaguchi H, Kimura F. Effects of glucose and related substrates on the recovery of the electrical activity of gonadotropin-releasing hormone pulse generator which is decreased by insulin-induced hypoglycemia in the estrogen- primed ovariectomized rat. Brain Res. 1999;820:71–76. doi: 10.1016/s0006-8993(98)01358-4. [DOI] [PubMed] [Google Scholar]
  • 101.Lu F, Yang JM, Wu JN, Chen YC, Kao YH, Tung CS, Yang SN. Activation of gonadotropin-releasing hormone receptors produces neuronal excitation in the rat hippocampus. Chin. J. Physiol. 1999;42:67–71. [PubMed] [Google Scholar]
  • 102.Osada T, Kimura F. LHRH effects on hippocampal-neurons are modulated by estrogen in rats. Endocr. J. 1995;42:251–257. doi: 10.1507/endocrj.42.251. [DOI] [PubMed] [Google Scholar]
  • 103.Gore AC, Windsor-Engnell BM, Terasawa E. Menopausal increases in pulsatile gonadotropin-releasing hormone release in a nonhuman primate (Macaca mulatta) Endocrinology. 2004;145:4653–4659. doi: 10.1210/en.2004-0379. [DOI] [PubMed] [Google Scholar]
  • 104.Atwood CS, Meethal SV, Liu T, Wilson AC, Gallego M, Smith MA, Bowen RL. Dysregulation of the hypothalamic-pituitary-gonadal axis with menopause and andropause promotes neurodegenerative senescence. J. Neuropathol. Exp. Neurol. 2005;64:93–103. doi: 10.1093/jnen/64.2.93. [DOI] [PubMed] [Google Scholar]
  • 105.Seong J, Park S, Kim K. Enhanced splicing of the first intron from the gonadotropin-releasing hormone (GnRH) primary transcript is a prerequisite for mature GnRH messenger RNA: presence of GnRH neuronspecific splicing factors. Mol. Endocrinol. 1999;13:1882–1895. doi: 10.1210/mend.13.11.0375. [DOI] [PubMed] [Google Scholar]
  • 106.Shim C, Khang I, Lee K-A, Kim K. Expression and regulation of gonadotropin- relasing hormone (GnRH) and its receptor mRNA transcripts during mouse ovarian development. Korean J. Biol. Sci. 2005;5:217–224. [Google Scholar]
  • 107.Jennes L, Woolums S. Localization of gonadotropin releasing hormone receptor mRNA in rat brain. Endocrine. 1994;2:521–528. [Google Scholar]
  • 108.Jennes L, Dalati B, Conn PM. Distribution of gonadotropin releasing hormone agonist binding sites in the rat central nervous system. Brain Res. 1988;452:156–164. doi: 10.1016/0006-8993(88)90020-0. [DOI] [PubMed] [Google Scholar]
  • 109.Renaud LP, Martin JB, Brazeau P. Depressant action of TRH, LH-RH and somatostatin on activity of central neurones. Nature. 1975;255:233–235. doi: 10.1038/255233a0. [DOI] [PubMed] [Google Scholar]
  • 110.Renaud LP, Martin JB, Brazeau P. Hypothalamic releasing factors: physiological evidence for a regulatory action on central neurons and pathways for their distribution in brain. Pharmacol. Biochem. Behav. 1976;5:171–178. doi: 10.1016/0091-3057(76)90348-8. [DOI] [PubMed] [Google Scholar]
  • 111.Quintanar JL, Salinas E. Neurotrophic effects of GnRH on neurite outgrowth and neurofilament protein expression in cultured cerebral cortical neurons of rat embryos. Neurochem. Res. 2008;33:1051–1056. doi: 10.1007/s11064-007-9549-9. [DOI] [PubMed] [Google Scholar]
  • 112.King JA, Millar RP. Identification of His5,Trp7,Tyr8-GnRH (chicken GnRH II) in amphibian brain. Peptides. 1986;7(5):827–834. doi: 10.1016/0196-9781(86)90102-6. [DOI] [PubMed] [Google Scholar]
  • 113.Maney DL, Richardson RD, Wingfield JC. Central administration of chicken gonadotropin-releasing hormone-II enhances courtship behavior in a female sparrow. Horm. Behav. 1997;32(1):11–18. doi: 10.1006/hbeh.1997.1399. [DOI] [PubMed] [Google Scholar]
  • 114.Temple JL, Millar RP, Rissman EF. An evolutionarily conserved form of gonadotropin-releasing hormone coordinates energy and reproductive behavior. Endocrinology. 2003;144(1):13–19. doi: 10.1210/en.2002-220883. [DOI] [PubMed] [Google Scholar]
  • 115.Jones SW. Chicken II luteinizing hormone-releasing hormone inhibits the M-current of bullfrog sympathetic neurons. Neurosci. Lett. 1987;80:180–184. doi: 10.1016/0304-3940(87)90650-1. [DOI] [PubMed] [Google Scholar]
  • 116.Darby S, Stockley J, Khan MM, Robson CN, Leung HY, Gnanapragasam VJ. Expression of GnRH type II is regulated by the androgen receptor in prostate cancer. Endocr. Relat. Cancer. 2007;14(3):613–624. doi: 10.1677/ERC-07-0041. [DOI] [PubMed] [Google Scholar]
  • 117.Emons G, Schröder B, Ortmann O, Westphalen S, Schulz KD, Schally AV. High affinity binding and direct antiproliferative effects of luteinizing hormone-releasing hormone analogs in human endometrial cancer cell lines. J. Clin. Endocrinol. Metab. 1993;77(6):1458–1464. doi: 10.1210/jcem.77.6.8263128. [DOI] [PubMed] [Google Scholar]
  • 118.Morgan K, Conklin D, Pawson AJ, Sellar R, Ott TR, Millar RP. A transcriptionally active human type II gonadotropin-releasing hormone receptor gene homolog overlaps two genes in the antisense orientation on chromosome 1q.12. Endocrinology. 2003;144:423–436. doi: 10.1210/en.2002-220622. [DOI] [PubMed] [Google Scholar]
  • 119.López de Maturana R, Pawson AJ, Lu ZL, Davidson L, Maudsley S, Morgan K, Langdon SP, Millar RP. Gonadotropin-releasing hormone analog structural determinants of selectivity for inhibition of cell growth: support for the concept of ligand-induced selective signaling. Mol. Endocrinol. 2008;22(7):1711–1722. doi: 10.1210/me.2006-0537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Cummings JL. Treatment of Alzheimer's disease: current and future therapeutic approaches. Rev. Neurol. Dis. 2004;1:60–69. [PubMed] [Google Scholar]
  • 121.Braak H, Rüb U, Schultz C, Del Tredici K. Vulnerability of cortical neurons to Alzheimer's and Parkinson's diseases. J. Alzheimers. Dis. 2006;9:35–44. doi: 10.3233/jad-2006-9s305. [DOI] [PubMed] [Google Scholar]
  • 122.Price JL, Davis PB, Morris JC, White DL. The distribution of tangles, plaques and related immunohistochemical markers in healthy aging and Alzheimer's disease. Neurobiol. Aging. 1991;12:295–312. doi: 10.1016/0197-4580(91)90006-6. [DOI] [PubMed] [Google Scholar]
  • 123.Kihara T, Shimohama S, Sawada H, Kimura J, Kume T, Kochiyama H, Maeda T, Akaike A. Nicotinic receptor stimulation protects neurons against beta-amyloid toxicity. Ann. Neurol. 1997;42:159–163. doi: 10.1002/ana.410420205. [DOI] [PubMed] [Google Scholar]
  • 124.Maudsley S, Mattson MP. Protein twists and turns in Alzheimer disease. Nat. Med. 2006;12:392–393. doi: 10.1038/nm0406-392. [DOI] [PubMed] [Google Scholar]
  • 125.Yao Y, Clark CM, Trojanowski JQ, Lee VM, Praticò D. Elevation of 12/15 lipoxygenase products in AD and mild cognitive impairment. Ann. Neurol. 2005;58:623–626. doi: 10.1002/ana.20558. [DOI] [PubMed] [Google Scholar]
  • 126.Hsiao K, Chapman P, Nilsen S, Eckman C, Harigaya Y, Younkin S, Yang F, Cole G. Correlative memory deficits, Abeta elevation, and amyloid plaques in transgenic mice. Science. 1996;274(5284):99–102. doi: 10.1126/science.274.5284.99. [DOI] [PubMed] [Google Scholar]
  • 127.van Groen T, Kiliaan AJ, Kadish I. Deposition of mouse amyloid beta in human APP/PS1 double and single AD model transgenic mice. Neurobiol. Dis. 2006;23(3):653–662. doi: 10.1016/j.nbd.2006.05.010. [DOI] [PubMed] [Google Scholar]
  • 128.Nelson RL, Guo Z, Halagappa VM, Pearson M, Gray AJ, Matsuoka Y, Brown M, Martin B, Iyun T, Maudsley S, Clark RF, Mattson MP. Prophylactic treatment with paroxetine ameliorates behavioral deficits and retards the development of amyloid and tau pathologies in 3xTgAD mice. Exp. Neurol. 2007;205:166–176. doi: 10.1016/j.expneurol.2007.01.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Martin B, Brenneman R, Becker KG, Gucek M, Cole RN, Maudsley S. iTRAQ analysis of complex proteome alterations in 3xTgAD Alzheimer’s mice: understanding the interface between physiology and disease. PLoS One. 2008;3(7):e2750. doi: 10.1371/journal.pone.0002750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Kumar-Singh S, Theuns J, Van Broeck B, Pirici D, Vennekens K, Corsmit E, Cruts M, Dermaut B, Wang R, Van Broeckhoven C. Mean age-of-onset of familial alzheimer disease caused by presenilin mutations correlates with both increased Abeta42 and decreased Abeta40. Hum. Mutat. 2006;27:686–695. doi: 10.1002/humu.20336. [DOI] [PubMed] [Google Scholar]
  • 131.Luheshi LM, Tartaglia GG, Brorsson AC, Pawar AP, Watson IE, Chiti F, Vendruscolo M, Lomas DA, Dobson CM, Crowther DC. Systematic in vivo analysis of the intrinsic determinants of amyloid Beta pathogenicity. PLoS Biol. 2007;5:e290. doi: 10.1371/journal.pbio.0050290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Roychaudhuri R, Yang M, Hoshi MM, Teplow DB. Amyloid beta-protein assembly and Alzheimer disease. J. Biol. Chem. 2009;284:4749–4753. doi: 10.1074/jbc.R800036200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Deshpande A, Mina E, Glabe C, Busciglio J. Different conformations of amyloid beta induce neurotoxicity by distinct mechanisms in human cortical neurons. J. Neurosci. 2006;26:6011–6018. doi: 10.1523/JNEUROSCI.1189-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Shankar GM, Li S, Mehta TH, Garcia-Munoz A, Shepardson NE, Smith I, Brett FM, Farrell MA, Rowan MJ, Lemere CA, Regan CM, Walsh DM, Sabatini BL, Selkoe DJ. Amyloid-beta protein dimers isolated directly from Alzheimer's brains impair synaptic plasticity and memory. Nat. Med. 2008;14:837–842. doi: 10.1038/nm1782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Selkoe DJ. Aging, amyloid, and Alzheimer's disease: a perspective in honor of Carl Cotman. Neurochem. Res. 2003;28(11):1705–1713. doi: 10.1023/a:1026065122854. [DOI] [PubMed] [Google Scholar]
  • 136.Selkoe DJ. Alzheimer's disease: genotypes, phenotypes, and treatments. Science. 1997;275:630–631. doi: 10.1126/science.275.5300.630. [DOI] [PubMed] [Google Scholar]
  • 137.Savonenko AV, Xu GM, Price DL, Borchelt DR, Markowska AL. Normal cognitive behavior in two distinct congenic lines of transgenic mice hyperexpressing mutant APPSWE. Neurobiol. Dis. 2003;12:194–211. doi: 10.1016/s0969-9961(02)00012-8. [DOI] [PubMed] [Google Scholar]
  • 138.Takeuchi A, Irizarry MC, Duff K, Saido TC, Hsiao Ashe K, Hasegawa M, Mann DM, Hyman BT, Iwatsubo T. Age-Related Amyloid β Deposition in Transgenic Mice Overexpressing Both Alzheimer Mutant Presenilin 1 and Amyloid β Precursor Protein Swedish Mutant Is Not Associated with Global Neuronal Loss. Am. J. Pathol. 2000;157:331–339. doi: 10.1016/s0002-9440(10)64544-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Perry G, Nunomura A, Raina AK, Smith MA. Amyloid-β junkies. Lancet. 2000;355:757. doi: 10.1016/S0140-6736(05)72173-5. [DOI] [PubMed] [Google Scholar]
  • 140.Obrenovich ME, Joseph JA, Atwood CS, Perry G, Smith MA. Alzheimer’s disease-a new beginning, or a final exit? In: Copani A, editor. Landes Bioscience. TX: NF; 2003. [Google Scholar]
  • 141.Rottkamp CA, Atwood CS, Joseph JA, Nunomura A, Perry G, Smith MA. The state versus amyloid-β: the trial of the most wanted criminal in Alzheimer disease. Peptides. 2002;23:1333–1341. doi: 10.1016/s0196-9781(02)00069-4. [DOI] [PubMed] [Google Scholar]
  • 142.Manly JJ, Merchant CA, Jacobs DM, Small SA, Bell K, Ferin M, Mayeux R. Endogenous estrogen levels and Alzheimer’s disease among postmenopausal women. Neurology. 2000;54:833–837. doi: 10.1212/wnl.54.4.833. [DOI] [PubMed] [Google Scholar]
  • 143.Rapp SR, Espeland MA, Shumaker SA, Henderson VW, Brunner RL, Manson JE, Gass ML, Stefanick ML, Lane DS, Hays J, Johnson KC, Coker LH, Dailey M, Bowen D. WHIMS Investigators. JAMA. 2003;289:2663–2672. doi: 10.1001/jama.289.20.2663. [DOI] [PubMed] [Google Scholar]
  • 144.Jennes L, Brame B, Centers A, Janovick JA, Conn PM. Regulation of hippocampal gonadotropin releasing hormone (GnRH) receptor mRNA and GnRH-stimulated inositol phosphate production by gonadal steroid hormones. Brain Res.Mol. Brain Res. 1995;33:104–110. doi: 10.1016/0169-328x(95)00113-7. [DOI] [PubMed] [Google Scholar]
  • 145.Thompson TL, Moss RL. Specific binding of 125ILHRH agonist to hippocampal membranes: fluctuations during the estrous cycle. Peptides. 1992;13:891–896. doi: 10.1016/0196-9781(92)90046-6. [DOI] [PubMed] [Google Scholar]
  • 146.Redding TW, Kastin AJ, Gonzales-Barcena D, Coy DH, Coy EJ, Schalch DS, Schally AV. The half-life, metabolism and excretion of tritiated luteinizing hormone-releasing hormone (LH-RH) in man. J. Clin. Endocrinol. Metab. 1973;37(4):626–631. doi: 10.1210/jcem-37-4-626. [DOI] [PubMed] [Google Scholar]
  • 147.Fauconnier JP, Teuwissen B, Thomas K. Rate of disappearance in plasma of synthetic LH-RH intravenously injected in man. Gynecol. Obstet. Invest. 1978;9(5):229–237. doi: 10.1159/000300989. [DOI] [PubMed] [Google Scholar]
  • 148.Bowen RL, Isley JP, Atkinson RL. An Association of Elevated Serum Gonadotropin Concentrations and Alzheimer Disease? J. Neuroendocrinol. 2000;12:351–354. doi: 10.1046/j.1365-2826.2000.00461.x. [DOI] [PubMed] [Google Scholar]
  • 149.Short SM, Boyer JL, Juliano RL. Integrins Regulate the Linkage between Upstream and Downstream Events in G Protein-coupled Receptor Signaling to Mitogen-activated Protein Kinase. J. Biol. Chem. 2000;275:12970–12977. doi: 10.1074/jbc.275.17.12970. [DOI] [PubMed] [Google Scholar]
  • 150.Yang SN, Lu F, Wu JN, Liu DD, Hsieh WY. Activation of gonadotropinreleasing hormone receptors induces a long-term enhancement of excitatory postsynaptic currents mediated by ionotropic glutamate receptors in the rat hippocampus. Neurosci. Lett. 1999;260:33–36. doi: 10.1016/s0304-3940(98)00939-2. [DOI] [PubMed] [Google Scholar]
  • 151.Prange-Kiel J, Jarry H, Schoen M, Kohlmann P, Lohse C, Zhou L, Rune GM. Gonadotropin-releasing hormone regulates spine density via its regulatory role in hippocampal estrogen synthesis. J. Cell. Biol. 2008;180:417–426. doi: 10.1083/jcb.200707043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Casadesus G, Webber KM, Atwood CS, Pappolla MA, Perry G, Bowen RL, Smith MA. Luteinizing hormone modulates cognition and amyloid-beta deposition in Alzheimer APP transgenic mice. Biochim. Biophys. Acta. 2006;1762(4):447–452. doi: 10.1016/j.bbadis.2006.01.008. [DOI] [PubMed] [Google Scholar]
  • 153.Vadakkadath Meethal S, Atwood CS. Alzheimer’s disease: the impact of age-related changes in reproductive hormones. Cell Mol. Life Sci. 2005;62(3):257–270. doi: 10.1007/s00018-004-4380-4. [DOI] [PubMed] [Google Scholar]
  • 154.Wang L, Martin B, Brenneman R, Luttrell LM, Maudsley S. Allosteric modulators of G protein-coupled receptors: future therapeutics for complex physiological disorders. J. Pharmacol.. Exp. Ther. 2009;331(2):340–348. doi: 10.1124/jpet.109.156380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.López de Maturana R, Martin B, Millar RP, Brown P, Davidson L, Pawson AJ, Nicol MR, Mason JI, Barran P, Naor Z, Maudsley S. GnRH-mediated DAN production regulates the transcription of the GnRH receptor in gonadotrope cells. Neuromolecular Med. 2007;9(3):230–248. doi: 10.1007/s12017-007-8004-z. [DOI] [PubMed] [Google Scholar]
  • 156.Maudsley S, Martin B, Luttrell LM. G protein-coupled receptor signaling complexity in neuronal tissue: implications for novel therapeutics. Curr. Alzheimer Res. 2007;4(1):3–19. doi: 10.2174/156720507779939850. [DOI] [PubMed] [Google Scholar]
  • 157.Martin B, Brenneman R, Golden E, Walent T, Becker KG, Prabhu VV, Wood W, 3rd, Ladenheim B, Cadet JL, Maudsley S. Growth factor signals in neural cells: coherent patterns of interaction control multiple levels of molecular and phenotypic responses. J. Biol. Chem. 2009;284(4):2493–2511. doi: 10.1074/jbc.M804545200. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES