Elsevier

Brain Research

Volume 1364, 10 December 2010, Pages 10-24
Brain Research

Review
Identified GnRH neuron electrophysiology: A decade of study

https://doi.org/10.1016/j.brainres.2010.09.066Get rights and content

Abstract

Over the past decade, the existence of transgenic mouse models in which reporter genes are expressed under the control of the gonadotropin-releasing hormone (GnRH) promoter has made possible the electrophysiological study of these cells. Here, we review the intrinsic and synaptic properties of these cells that have been revealed by these approaches, with a particular regard to burst generation. Advances in our understanding of neuromodulation of GnRH neurons and synchronization of this network are also discussed.

Introduction

The gonadotropin-releasing hormone (GnRH) neuronal system is the final common pathway for central regulation of fertility. Through its regulation of the synthesis and secretion of the pituitary gonadotropins luteinizing hormone (LH, often used as a surrogate marker for GnRH release) and follicle-stimulating hormone (FSH), GnRH exerts control over the hypothalamo–pituitary gonadal axis. Multiple inputs from the internal environment (e.g., steroid hormones, energy balance, stress) and external environment (e.g., photoperiod, pheromones, endocrine disruptors) can alter fertility through central actions that ultimately alter the release of GnRH. Even before the sequencing of the GnRH decapeptide (Baba et al., 1971), both the innate function of this system and its response to various stimuli were intensely studied. During much of this time, the ability to tease out the neurobiological mechanisms underlying observations of altered GnRH or LH release was limited by the technical approaches available and the scattered distribution of GnRH neurons. It was possible, for example, to determine if a factor acted centrally by intracerebroventricular application or treatment of hypothalamic explants or cultures and to speculate about upstream afferents through the localization of steroid receptors or expression of the immediate early gene cFos (Hoffman et al., 1990, Lehman and Karsch, 1993, Shivers et al., 1983), knife cut studies (Blake and Sawyer, 1974) or localized administration of steroids (McManus et al., 2005). But understanding the intrinsic, synaptic and network properties of these neurons was very limited in mammalian systems. In contrast, electrophysiological work in the teleost dwarf gourami provided considerable insight into pacemaker activities of the terminal nerve GnRH neurons, which can be easily identified by the eye (Oka, 1995, Oka and Matsushima, 1993).

Despite the limitations largely precluding direct study of mammalian GnRH neurons, a great deal of knowledge about the reproductive neuroendocrine system was acquired and has served as the basis for designing and interpreting experiments that are now taking advantage of new model systems. The pioneering work of Ernst Knobil's group describing circhoral oscillations of LH in the circulation (Dierschke et al., 1970), the importance of episodic GnRH stimulation for pituitary function (Belchetz et al., 1978, Wildt et al., 1981) and central electrical correlates with LH release (Wilson et al., 1984) still provides the fundamental description of this system as a pulse generator and drives many current research questions. Several groups independently developed methods to measure directly GnRH release (Caraty and Locatelli, 1988, Clarke and Cummins, 1982, Levine et al., 1985, Levine and Ramirez, 1980). Both immortalized GnRH neuron cell lines (Mellon et al., 1990, Radovick et al., 1991) and primary embryonic cultures derived from the migratory pathway of GnRH neurons first in the primate (Terasawa et al., 1993) and later in the mouse (Fueshko and Wray, 1994) also provided insight into the function of this system. None of these approaches, however, allowed examination of native adult GnRH neurons in the context of at least a portion of their normal network using the powerful electrophysiological methods that were being applied to other central neuronal cell types that could be identified by anatomical location.

Martin Kelly's group was the first to study individual GnRH neurons using electrophysiological recordings in acutely prepared brain slices, demonstrating that GnRH neurons were acutely hyperpolarized by estradiol in the guinea pig model (Kelly et al., 1984). This and subsequent work in this model (Lagrange et al., 1995, Wagner et al., 1998) identified the first intrinsic properties of mammalian GnRH neurons. The effort needed to accumulate GnRH neurons, however, was heroic; in the first study, which utilized Procion yellow labeling during blind sharp electrode recordings followed by post hoc identification, only 5 of 102 cells studied were GnRH positive on post hoc examination.

About a decade ago, promoter-driven reporter genes were applied to the identification of GnRH neurons. The cell-specifically and strongly expressed GnRH promoter is well suited to this approach and was used to drive expression of modified jellyfish Aequorea victoria green fluorescent protein (GFP), beta galactosidease or the calcium indicator Pericam (Han et al., 2004, Jasoni et al., 2007, Kanda et al., 2010, Kato et al., 2003, Skynner et al., 1999, Spergel et al., 1999, Suter et al., 2000a, Wayne et al., 2005) in GnRH neurons from mouse, rat and medaka. Primarily using the GFP models, considerable progress has been made over the past decade in the understanding of the neurobiology of GnRH neurons. These studies will form the basis of this review. The reader is also pointed to a recent excellent review on the intracellular calcium dynamics in GnRH neurons (Jasoni et al., In press) and previous reviews on the physiology of the GnRH neuronal system (Herbison, 2006, Moenter et al., 2003).

Section snippets

An overview of electrophysiological approaches used in the GnRH system

The purpose of this review is not to provide a complete primer on electrophysiological approaches, which are available elsewhere (Hille, 2001, Sakman and Neher, 1995). What follows is a brief overview of the different approaches that have been used on GnRH neurons, their plusses and minuses and the types of data that can be acquired with each.

Intrinsic properties of GnRH neurons

Observations of the episodic activity of multiunit electrical activity and hormone release at the whole animal level (Moenter et al., 1992, Wilson et al., 1984) coupled with the electrophysiology magnocellular neuroendocrine neurons (Andrew and Dudek, 1983, Dutton and Dyball, 1979, Dutton et al., 1978, Nordmann and Stuenkel, 1986, Wakerley et al., 1978, Wakerley et al., 1975) showing that action potential firing, in particular burst firing, was needed for hormone release, generated several

Steroid modulation of GnRH neuron firing pattern and underlying conductances

A critical aspect of the GnRH neuronal system is its feedback regulation by steroid hormones. GnRH neurons express the beta isoform of the estradiol receptor (Herbison and Pape, 2001, Hrabovszky et al., 2000, Hrabovszky et al., 2001), but most other steroid receptors have only rarely, if at all, been detected in GnRH neurons. This has led to an overall view in the field that steroid regulation of GnRH neurons engages steroid-sensitive afferent neurons (Wintermantel et al., 2006). Direct

Neurotransmitters, neuromodulators and GnRH neuron function

When discussing either acute or chronic effects of steroids on GnRH neurons, the likelihood of intermediate neurons being involved necessitates an examination of these factors. The primary conveyors of fast synaptic transmission (mediated by ionotropic vs. metabotropic receptors) are GABA and glutamate. GnRH neurons receive spontaneous glutamatergic transmission via both AMPA/KA and NMDA receptors (Chen and Moenter, 2009, Christian et al., 2009, Spergel et al., 1999, Suter, 2004), but the

Coordination of GnRH neurons

The episodic nature of GnRH release mandates some level of coordination among these cells. Studies in GT1 cells suggested that not every cell participates in every burst of activity (Nunemaker et al., 2001). Similarly, oscillations in intracellular calcium levels indicate coordination among a part of the population (Abe et al., 2008). Unfortunately, relatively little experimental evidence can positively support a mechanism for how the GnRH neuronal network produces pulses, however some findings

Summary

The past decade has seen a major increase in our knowledge of the intrinsic and synaptic properties of GnRH neurons. The expanding repertoire of intrinsic and synaptic properties can continue to be incorporated into sophisticated models that generate new hypotheses to be tested about the network. Questions we are now poised to address include the following: What are the roles of non-GnRH neuron and non-neuronal elements in coordinating the network? How do the various conductances that

References (165)

  • P.L. Mellon et al.

    Immortalization of hypothalamic GnRH neurons by genetically targeted tumorigenesis

    Neuron

    (1990)
  • S.M. Moenter et al.

    Mechanisms underlying episodic gonadotropin-releasing hormone secretion

    Front. Neuroendocrinol.

    (2003)
  • H. Abe et al.

    Mechanisms of the modulation of pacemaker activity by GnRH peptides in the terminal nerve-GnRH neurons

    Zool. Sci.

    (2002)
  • H. Abe et al.

    Firing pattern and rapid modulation of activity by estrogen in primate luteinizing hormone releasing hormone-1 neurons

    Endocrinology

    (2005)
  • H. Abe et al.

    Rapid action of estrogens on intracellular calcium oscillations in primate LHRH-1 neurons

    Endocrinology

    (2008)
  • Y. Akazome et al.

    Functional and evolutionary insights into vertebrate kisspeptin systems from studies of fish brain

    J. Fish Biol.

    (2010)
  • R.D. Andrew et al.

    Burst discharge in mammalian neuroendocrine cells involves an intrinsic regenerative mechanism

    Science

    (1983)
  • P. Belchetz et al.

    Hypophysial responses to continuous and intermittent delivery of hypothalamic gonadotropin-releasing hormone

    Science

    (1978)
  • Bhattarai, J.P., Kaszas, A., Park, S.A., Yin, H., Park, S.J., Herbison, A.E., Han, S.-K. and Abraham, I.M., In press....
  • C.A. Blake et al.

    Effects of hypothalamic deafferentation on the pulsatile rhythm in plasma concentrations of luteinizing hormone in ovariectomized rats

    Endocrinology

    (1974)
  • M.A. Bosch et al.

    Distribution, neuronal colocalization, and 17beta-E2 modulation of small conductance calcium-activated K(+) channel (SK3) mRNA in guinea pig brain

    Endocrinology

    (2002)
  • R.E. Campbell et al.

    Dendro–dendritic bundling and shared synapses between gonadotropin-releasing hormone neurons

    Proc. Natl Acad. Sci. USA

    (2009)
  • A. Caraty et al.

    Effect of time after castration on secretion of LHRH and LH in the ram

    J. Reprod. Fertil.

    (1988)
  • Y.M. Chan et al.

    Kisspeptin/Gpr54-independent gonadotrophin-releasing hormone activity in Kiss1 and Gpr54 mutant mice

    J. Neuroendocrinol.

    (2009)
  • A.C. Charles et al.

    Mechanisms of spontaneous calcium oscillations and action potentials in immortalized hypothalamic (GT1-7) neurons

    J. Neurophysiol.

    (1995)
  • P. Chen et al.

    GABAergic transmission to gonadotropin-releasing hormone (GnRH) neurons is regulated by GnRH in a concentration-dependent manner engaging multiple signaling pathways

    J. Neurosci.

    (2009)
  • C.A. Christian et al.

    Estradiol induces diurnal shifts in GABA transmission to gonadotropin-releasing hormone neurons to provide a neural signal for ovulation

    J. Neurosci.

    (2007)
  • C.A. Christian et al.

    Vasoactive intestinal polypeptide can excite gonadotropin-releasing hormone neurons in a manner dependent on estradiol and gated by time of day

    Endocrinology

    (2008)
  • Christian, C.A. and Moenter, S.M., In press. The neurobiology of preovulatory and estradiol-induced...
  • C.A. Christian et al.

    Diurnal and estradiol-dependent changes in gonadotropin-releasing hormone neuron firing activity

    Proc. Natl Acad. Sci. USA

    (2005)
  • C.A. Christian et al.

    Classical estrogen receptor alpha signaling mediates negative and positive feedback on gonadotropin-releasing hormone neuron firing

    Endocrinology

    (2008)
  • C.A. Christian et al.

    Estradiol suppresses glutamatergic transmission to gonadotropin-releasing hormone neurons in a model of negative feedback in mice

    Biol. Reprod.

    (2009)
  • Z. Chu et al.

    Endogenous activation of metabotropic glutamate receptors modulates GABAergic transmission to gonadotropin-releasing hormone neurons and alters their firing rate: a possible local feedback circuit

    J. Neurosci.

    (2005)
  • Z. Chu et al.

    Physiologic regulation of a tetrodotoxin-sensitive sodium influx that mediates a slow afterdepolarization potential in gonadotropin-releasing hormone neurons: possible implications for the central regulation of fertility

    J. Neurosci.

    (2006)
  • Z. Chu et al.

    Acute estradiol alters gonadotropin-releasing hormone (GnRH) neuron excitability

    Soc. Neurosci. Abstr. Viewer

    (2007)
  • Z. Chu et al.

    Differential regulation of gonadotropin-releasing hormone neuron activity and membrane properties by acutely applied estradiol: dependence on dose and estrogen receptor subtype

    J. Neurosci.

    (2009)
  • Z. Chu et al.

    Hyperpolarization-activated currents in gonadotropin-releasing hormone (GnRH) neurons contribute to intrinsic excitability and are regulated by gonadal steroid feedback

    J. Neurosci.

    (2010)
  • I.J. Clarke et al.

    The temporal relationship between gonadotropin releasing hormone (GnRH) and luteinizing hormone (LH) secretion in ovariectomized ewes

    Endocrinology

    (1982)
  • Clarkson, J., Han, S.-K., Liu, X., Lee, K. and Herbison, A.E., In press. Neurobiological mechanisms underlying...
  • N. de Roux et al.

    Hypogonadotropic hypogonadism due to loss of function of the KiSS1-derived peptide receptor GPR54

    Proc. Natl Acad. Sci. USA

    (2003)
  • R.A. DeFazio et al.

    Estradiol feedback alters potassium currents and firing properties of gonadotropin-releasing hormone neurons

    Mol. Endocrinol.

    (2002)
  • R.A. DeFazio et al.

    Activation of A-type gamma-aminobutyric acid receptors excites gonadotropin-releasing hormone neurons

    Mol. Endocrinol.

    (2002)
  • L.V. DePaolo et al.

    In vivo and in vitro examination of an autoregulatory mechanism for luteinizing hormone-releasing hormone

    Endocrinology

    (1987)
  • D.J. Dierschke et al.

    Circhoral oscillations of plasma LH levels in the ovariectomized rhesus monkey

    Endocrinology

    (1970)
  • F. Docke et al.

    The mechanism of the induction of ovulation by oestrogens

    J. Endocrinol.

    (1965)
  • E. Ducret et al.

    RFamide-related peptide-3, a mammalian gonadotropin-inhibitory hormone ortholog, regulates gonadotropin-releasing hormone neuron firing in the mouse

    Endocrinology

    (2009)
  • I. Dumalska et al.

    Excitatory effects of the puberty-initiating peptide kisspeptin and group I metabotropic glutamate receptor agonists differentiate two distinct subpopulations of gonadotropin-releasing hormone neurons

    J. Neurosci.

    (2008)
  • A. Dutton et al.

    Phasic firing enhances vasopressin release from the rat neurohypophysis

    J. Physiol.

    (1979)
  • A. Dutton et al.

    The importance of short interpulse intervals in determining vasopressin release from the isolated neurohypophysis [proceedings]

    J. Physiol.

    (1978)
  • E.S. Faber et al.

    Physiological role of calcium-activated potassium currents in the rat lateral amygdala

    J. Neurosci.

    (2002)
  • Cited by (54)

    • Modelling KNDy neurons and gonadotropin-releasing hormone pulse generation

      2022, Current Opinion in Endocrine and Metabolic Research
      Citation Excerpt :

      The GnRH neuron is an integral part of the pulse generator as it secretes GnRH to the pituitary; however, this does not necessarily mean the generator is solely located within the GnRH neuronal population. As mentioned above, the pulsatile nature of GnRH release can be externally modulated or even wholly driven by upstream inputs, with the obvious possibility being stimulation by kisspeptin [52]. Given that kisspeptin acts directly on GnRH neurons [53,54] and is a strong candidate for influencing GnRH release, the model previously developed by Chen et al. [17] was expanded to account for the impact of kisspeptin on the behaviour of the GnRH neuron [55].

    • Unraveling the Neural Mechanisms Underlying the GnRH Pulse Generator: An Update

      2021, Cellular Endocrinology in Health and Disease, Second Edition
    • A simple model of estrous cycle negative and positive feedback regulation of GnRH secretion

      2020, Frontiers in Neuroendocrinology
      Citation Excerpt :

      Implicit in all of these models was the assumption that the ultimate pattern of GnRH secretion results from neural integration of various afferent pathways mediating the positive and negative feedback signals at the GnRH neuron cell body. As such, very many studies have compared the morphological and electrical properties of the GnRH neuron soma during conditions simulating estrogen negative and positive feedback (Moenter 2010, Herbison 2015). This mini-review first describes the proposed model and then provides a biological basis for its components and operation.

    • Timing reproduction in teleost fish: Cues and mechanisms

      2016, Current Opinion in Neurobiology
      Citation Excerpt :

      Gravid females respond to the courtship hum of the males, and their sensitivity to the frequency range of the hum increases during the summer when they lay eggs. Across all vertebrates, GnRH1 neurons exhibit coordinated, pulsatile activity, which is postulated to overcome a signaling threshold in the pituitary [35•]. Gonadotropin-producing cells of the pituitary rapidly desensitize to tonic levels of GnRH1 [36••].

    • Gonadotropin-releasing hormone neurons

      2016, The Curated Reference Collection in Neuroscience and Biobehavioral Psychology
    View all citing articles on Scopus
    View full text