The KiNG of reproduction: Kisspeptin/ nNOS interactions shaping hypothalamic GnRH release

https://doi.org/10.1016/j.mce.2021.111302Get rights and content

Highlights

  • nNOS neurons use local signaling when their activity is sparse or uncoordinated, and volume signaling when synchronous.

  • Kisspeptin can promote nNOS phosphorylation, enabling nNOS synchronous activity and subsequently NO release.

  • NO has opposite effects to kisspeptin on GnRH secretion.

  • NO acts as a brake allowing GnRH neurons to better synchronize, priming them for the excitatory kisspeptin stimulus.

  • NO and kisspeptin, by acting as the Yin and Yang of the GnRH axis may shape GnRH pulses and surges.

Abstract

Gonadotropin-releasing hormone (GnRH) is the master regulator of the hypothalamic-pituitary-gonadal (HPG) axis, and therefore of fertility and reproduction. The release pattern of GnRH by the hypothalamus includes both pulses and surges. However, despite a considerable body of evidence in support of a determinant role for kisspeptin, the mechanisms regulating a GnRH pulse and surge remain a topic of debate. In this review we challenge the view of kisspeptin as an absolute “monarch”, and instead present the idea of a Kisspeptin-nNOS-GnRH or “KiNG” network that is responsible for generating the “GnRH pulse” and “GnRH surge”. In particular, the neuromodulator nitric oxide (NO) has opposite effects to kisspeptin on GnRH secretion in many respects, acting as the Yin to kisspeptin's Yang and creating a dynamic system in which kisspeptin provides the “ON” signal, promoting GnRH release, while NO mediates the “OFF” signal, acting as a tonic brake on GnRH secretion. This interplay between an activator and an inhibitor, which is in turn fine-tuned by the gonadal steroid environment, thus leads to the generation of GnRH pulses and surges and is crucial for the proper development and function of the reproductive axis.

Introduction

ὁρμῶν (hormôn), a Greek word meaning “to set in motion, to excite, to stimulate”. Since its introduction by Ernest Starling in 1905 (Starling, 1905), this word has been used to describe the chemical messengers used as a means of communication between different organs in an animal. When the term was introduced, practically nothing was known about the nature or the action of these messengers, which were believed to be produced by only a few specialized organs of the endocrine system (i.e. the glands). We have come a long way since then, with many conceptual changes occurring over the years, the most important of which is possibly the acknowledgement that the nervous and endocrine systems work together to transmit physiological information. The discipline of “Neuroendocrinology” was launched by Geoffrey Harris with his publication in 1955, which not only provided the first proof that the endocrine system could be controlled by the central nervous system (CNS), but also laid the foundations for the notion of the hypothalamic-pituitary-gonadal axis (HPG) (Harris, 1955). In the past two decades, we have come to acknowledge that the field of neuroendocrinology extends far beyond the traditional neuron-endocrine pathways to encompass the production of hormones by non-traditional cells and tissues, with new and often non-catalytic roles in the regulation of an organism's development, physiological homeostasis, reproductive capacity and behavior.

The three components of the HPG axis – the hypothalamus, pituitary gland and gonads (i.e. the testes and ovaries) – closely interact and depend on each other to allow the complex dialogue between the CNS and the periphery that is indispensable for reproductive function. The hypothalamus is undeniably the single most important brain region integrating vegetative and endocrine signals, and controls diverse processes including cardiovascular function, sleep, metabolism, stress, thermoregulation, water and electrolyte balance, growth and reproduction. Within the hypothalamus, specialized neuronal populations sense moment-to-moment changes in circulating levels of hormones and nutrients, to regulate neuroendocrine function (Elmquist et al., 2005). Among these hypothalamic neuronal populations are the neurons producing gonadotropin-releasing hormone (GnRH), the main orchestrators of reproductive function, which act as integrators of various signals coming from both the central and the peripheral nervous system.

In spite of their crucial role, GnRH neurons are an extremely small population of cells across mammalian species, counting only 1000–3000 neurons in the rodent brain. In rodents, the GnRH neuronal soma are primarily distributed in the preoptic hypothalamic area (POA) extending their nerve terminals to the pericapillary space of the median eminence (ME), located in the more mediobasal area of the hypothalamus (MBH) (Barry J et al., 1973), releasing the GnRH decapeptide in an episodic manner in both sexes (Sarkar et al., 1976; Moenter et al., 1991; Sarkar and Minami, 1995; Terasawa et al., 1999). Indeed, both immortalized GnRH-secreting GT1 cells and primary GnRH neurons release GnRH in a pulsatile manner, at species-specific intervals (for review see Terasawa, 2019). GnRH is then carried through the pituitary portal circulation for delivery to the anterior pituitary, where it stimulates gonadotropes to synthesize and secrete the gonadotropins, luteinizing hormone (LH) and follicle stimulating hormone (FSH). These gonadotropins then act on the gonads (i.e. the testes and ovaries) to promote gonadal development and the secretion of sex steroids, which in turn provide positive or negative feedback back to the brain to regulate GnRH release (Prevot, 2015). The pulsatile pattern of GnRH secretion is reflected in the pulsatile secretion of LH during the negative feedback action of sex steroids, while GnRH surge, taking place during the positive feedback action of sex steroids, results in a crucial peak of LH release, triggering ovulation in females (Nett et al., 1974).

Thus, correct release of LH and FSH depends on the proper regulation of the frequency and timing of GnRH secretion by sex steroids as well as other neuronal and non-neuronal factors. In turn, proper development of GnRH neurons, GnRH expression and GnRH signaling are all essential for normal functioning of the mammalian HPG axis (Cattanach et al., 1977; Mason et al., 1986; Schwanzel-Fukuda et al., 1989). Justified by the authority of the GnRH system over the regulation of key physiological events, the network surrounding GnRH neurons, ensuring the controlled and timely regulation of their response is complex and multidimensional. Among these cells, kisspeptin neurons have so far been considered the master excitatory driving force behind GnRH/LH release during both positive and negative feedback phases (Navarro et al., 2009; Pielecka-Fortuna et al., 2010; Clarkson et al., 2017).

In this review, based primarily on research carried out in rodents, we challenge the view of kisspeptin neurons as the sole regulators or supreme “monarchs” of the GnRH network, controlling both GnRH pulse and surge generation. Instead, we explore the implication of the much overlooked population of neuronal nitric oxide synthase (nNOS) neurons, producing the diffusible messenger nitric oxide (NO), in the control of the GnRH system. Finally, we will discuss how the tripartite Kisspeptin, nNOS, GnRH (KiNG) network, through a mechanism consisting of the alternation of neuronal activation and the release of tonic inhibition, regulates LH pulsatility and LH surge generation and thus reproductive function.

Section snippets

Milestones during the developmental maturation of the GnRH network in the mouse

GnRH neurons originate from stem cells of the olfactory placode. Around embryonic (E) day 11.5 in mice, these neurons embark on their migratory path from the nose, entering the forebrain following a series of guidance cues (for review see Wray et al., 2010), and in close association with the vomeronasal nerve fibers. By E16, GnRH neurons enter the forebrain (Schwanzel-Fukuda et al., 1989). The HPG axis is believed to be already somewhat functional during embryonic development (Aubert et al.,

The involvement of ovarian steroid hormones in the GnRH pulse and the GnRH surge generators

Even though the secretory profiles of estrogen and progesterone are poorly characterized in mice, both gonadal steroids exert critical inhibitory and stimulatory actions upon the brain to control GnRH release, shaping the estrous cycle in all female mammals.

It is widely accepted that estrogens play a key role, exerting a negative feedback action on the GnRH system, thus maintaining the constant frequency of the episodic release of GnRH/LH throughout the follicular phase. According to the

The mechanism underlying the ability of nNOS cells to promote the synchronized activity of GnRH neurons

Since mammalian GnRH neurons are widely scattered in the POA, one could hypothesize that GnRH pulsatility and GnRH surge are dependent upon a form of oscillatory network activity rather than individual synaptic connectivity. Although this concept remains to be uncovered in vivo, this oscillatory activity may ultimately reflect the wave-like shape of both pulsatile and surge GnRH secretion. Considering that nNOS neurons can communicate with each other via NO volume transmission, NO signaling is

Concluding remarks and perspectives

Overall, the kisspeptin/nNOS neuronal network is ideally poised to generate and regulate episodic GnRH release in response to developmental and physiological cues, thus ensuring the precise sequence of events that constitutes pubertal activation and adult fertility (Choe et al., 2013; Messina et al., 2016). However, upstream of its release, GnRH must also be produced. Recent findings shed some light on the transcriptional mechanisms controlling the postnatal maturation of the GnRH system,

Acknowledgements

The authors acknowledge S. Rasika for editorial assistance with the manuscript before submission. The author's work is supported by a doctoral fellowship from the University of Lille School of Medicine, Lille, France (to V.D.), the Fondation pour la Recherche Médicale (Equipe FRM, DEQ20130326524 to V.P) and the Agence Nationale de la Recherche (ANR-17-CE16-0015 to V.P) and the European Union Horizon 2020 research and innovation program (No 847941 to K.C. and V.P.).

References (163)

  • M. Kotani et al.

    The metastasis suppressor gene KiSS-1 encodes kisspeptins, the natural ligands of the orphan G protein-coupled receptor GPR54

    J. Biol. Chem.

    (2001)
  • D.K. Lee et al.

    Discovery of a receptor related to the galanin receptors

    FEBS Lett.

    (1999)
  • M.N. Lehman et al.

    Importance of neuroanatomical data from domestic animals to the development and testing of the KNDy hypothesis for GnRH pulse generation

    Domest. Anim. Endocrinol.

    (2020)
  • J.E. Levine

    Chapter 26—neuroendocrine control of the ovarian cycle of the rat

  • A.P. Abreu et al.

    Central precocious puberty caused by mutations in the imprinted gene MKRN3

    N. Engl. J. Med.

    (2013)
  • A.P. Abreu et al.

    MKRN3 inhibits the reproductive axis through actions in kisspeptin-expressing neurons

    J. Clin. Invest.

    (2020)
  • M. Amstalden et al.

    Neurokinin 3 receptor immunoreactivity in the septal region, preoptic area and hypothalamus of the female sheep: colocalisation in neurokinin B cells of the arcuate nucleus but not in gonadotrophin-releasing hormone neurones

    J. Neuroendocrinol.

    (2010)
  • M.L. Aubert et al.

    Ontogeny of hypothalamic luteinizing hormone-releasing hormone (GnRH) and pituitary GnRH receptors in fetal and neonatal rats

    Endocrinology

    (1985)
  • Barry J et al.

    LRF producing cells of the mammalian hypothalamus. A fluorescent antibody study

    Zeitschrift fur Zellforschung und mikroskopische Anatomie

    (1973)
  • A.M. Batchelor et al.

    Exquisite sensitivity to subsecond, picomolar nitric oxide transients conferred on cells by guanylyl cyclase-coupled receptors

    Proc. Natl. Acad. Sci. Unit. States Am.

    (2010)
  • M.N. Bedenbaugh et al.

    Pubertal escape from estradiol negative feedback in Ewe lambs is not accounted for by decreased ESR1 mRNA or protein in kisspeptin neurons

    Endocrinology

    (2018)
  • M.N. Bedenbaugh et al.

    Neuroanatomical relationship of neuronal nitric oxide synthase to gonadotropin-releasing hormone and kisspeptin neurons in adult female sheep and primates

    Neuroendocrinology

    (2018)
  • M.N. Bedenbaugh et al.

    Kisspeptin, gonadotrophin-releasing hormone and oestrogen receptor α colocalise with neuronal nitric oxide synthase neurones in prepubertal female sheep

    J. Neuroendocrinol.

    (2018)
  • N. Bellefontaine et al.

    Nitric oxide as key mediator of neuron-to-neuron and endothelia-to-glia communication involved in the neuroendocrine control of reproduction

    Neuroendocrinology

    (2011)
  • N. Bellefontaine et al.

    Leptin-dependent neuronal NO signaling in the preoptic hypothalamus facilitates reproduction

    J. Clin. Invest.

    (2014)
  • J.J. Bonavera et al.

    Evidence that nitric oxide may mediate the ovarian steroid-induced luteinizing hormone surge: involvement of excitatory amino acids

    Endocrinology

    (1993)
  • S.G. Bouret et al.

    Trophic action of leptin on hypothalamic neurons that regulate feeding

    Science

    (2004)
  • A. Caraty et al.

    Evidence that the mediobasal hypothalamus is the primary site of action of estradiol in inducing the preovulatory gonadotropin releasing hormone surge in the Ewe

    Endocrinology

    (1998)
  • M.-P. Caron et al.

    Dual-acting riboswitch control of translation initiation and mRNA decay

    Proc. Natl. Acad. Sci. Unit. States Am.

    (2012)
  • B.M. Cattanach et al.

    Gonadotrophin-releasing hormone deficiency in a mutant mouse with hypogonadism

    Nature

    (1977)
  • K. Chachlaki et al.

    The gentle art of saying NO: how nitric oxide gets things done in the hypothalamus

    Nat. Rev. Endocrinol.

    (2017)
  • K. Chachlaki et al.

    Phenotyping of nNOS neurons in the postnatal and adult female mouse hypothalamus

    J. Comp. Neurol.

    (2017)
  • K. Chachlaki et al.

    Nitric oxide signalling in the brain and its control of bodily functions

    Br. J. Pharmacol.

    (2020)
  • R.Y. Cheong et al.

    Expression of ESR1 in glutamatergic and GABAergic neurons is essential for normal puberty onset, estrogen feedback, and fertility in female mice

    J. Neurosci.

    (2015)
  • H.K. Choe et al.

    Synchronous activation of gonadotropin-releasing hormone gene transcription and secretion by pulsatile kisspeptin stimulation

    Proc. Natl. Acad. Sci. Unit. States Am.

    (2013)
  • C.A. Christian et al.

    The neurobiology of preovulatory and estradiol-induced gonadotropin-releasing hormone surges

    Endocr. Rev.

    (2010)
  • J. Clarkson et al.

    Postnatal development of kisspeptin neurons in mouse hypothalamus; sexual dimorphism and projections to gonadotropin-releasing hormone neurons

    Endocrinology

    (2006)
  • J. Clarkson et al.

    Kisspeptin-GPR54 signaling is essential for preovulatory gonadotropin-releasing hormone neuron activation and the luteinizing hormone surge

    J. Neurosci.: Off. J. Soc. Neurosci.

    (2008)
  • J. Clarkson et al.

    Definition of the hypothalamic GnRH pulse generator in mice

    Proc. Natl. Acad. Sci. Unit. States Am.

    (2017)
  • J. Clasadonte et al.

    Activation of neuronal nitric oxide release inhibits spontaneous firing in adult gonadotropin-releasing hormone neurons: a possible local synchronizing signal

    Endocrinology

    (2008)
  • S. Constantin et al.

    Nitric oxide resets kisspeptin-excited GnRH neurons via PIP2 replenishment

    Proc. Natl. Acad. Sci. Unit. States Am.

    (2021)
  • E.C. Cottrell et al.

    Postnatal remodeling of dendritic structure and spine density in gonadotropin-releasing hormone neurons

    Endocrinology

    (2006)
  • X. d'Anglemont de Tassigny et al.

    Coupling of neuronal nitric oxide synthase to NMDA receptors via postsynaptic density-95 depends on estrogen and contributes to the central control of adult female reproduction

    J. Neurosci.

    (2007)
  • X. d'Anglemont de Tassigny et al.

    Estradiol induces physical association of neuronal nitric oxide synthase with NMDA receptor and promotes nitric oxide formation via estrogen receptor activation in primary neuronal cultures

    J. Neurochem.

    (2009)
  • X. d'Anglemont de Tassigny et al.

    The role of kisspeptin signaling in reproduction

    Physiology

    (2010)
  • X. d'Anglemont de Tassigny et al.

    Kisspeptin signaling is required for peripheral but not central stimulation of gonadotropin-releasing hormone neurons by NMDA

    J. Neurosci. : Off. J. Soc. Neurosci.

    (2010)
  • S. De Seranno et al.

    Vascular endothelial cells promote acute plasticity in ependymoglial cells of the neuroendocrine brain

    J. Neurosci.

    (2004)
  • N. de Roux et al.

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

    Proc. Natl. Acad. Sci. U.S.A.

    (2003)
  • S.L. Dubois et al.

    Positive, but not negative feedback actions of estradiol in adult female mice require estrogen receptor α in kisspeptin neurons

    Endocrinology

    (2015)
  • J.K. Elmquist et al.

    Identifying hypothalamic pathways controlling food intake, body weight, and glucose homeostasis

    J. Comp. Neurol.

    (2005)
  • Cited by (0)

    View full text