1
|
Dorfman VB. Distribution of the kisspeptin system and its relation with gonadotropin-releasing hormone in the hypothalamus. VITAMINS AND HORMONES 2024; 127:51-78. [PMID: 39864946 DOI: 10.1016/bs.vh.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Kisspeptin (KISS1), originally catalogued as metastin because of its capacity as a metastasis suppressor in human melanoma and breast cancer, is now recognized as the major puberty gatekeeper and gonadotropin-releasing hormone (GnRH) neuroendocrine system modulator. It is a member of the family of RFamide-related peptides that also includes the neuropeptide FF group, the gonadotropin-inhibitory hormone, the prolactin-releasing peptide, and the 26RFa peptides. The KISS1 precursor peptide is processed into a family of peptides known as kisspeptins. Its expression has been described in the hypothalamus as well as in the whole reproductive axis and several extra reproductive tissues of mammals as well as fish and amphibians, but not in birds. KISS1 plays an essential role as a regulator of the reproductive axis by inducing the synthesis and release of GnRH, acting through specific receptors. The study of the kisspeptin system and its relation with reproduction in wild and non-classical laboratory species is extremely useful to understand and become aware of the role of KISS1 in the wide variety of possible different reproductive strategies. In this chapter, KISS1 involvement in non-classical laboratory rodents, fishes, and birds is discussed.
Collapse
Affiliation(s)
- Verónica Berta Dorfman
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina.
| |
Collapse
|
2
|
Panda SP, Kesharwani A, Singh GD, Prasanth D, Vatchavai BR, Kumari PVK, Panda SK, Mallick SP. Impose of KNDy/GnRH neural circuit in PCOS, ageing, cancer and Alzheimer's disease: StAR actions in prevention of neuroendocrine dysfunction. Ageing Res Rev 2023; 92:102086. [PMID: 37821047 DOI: 10.1016/j.arr.2023.102086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/06/2023] [Accepted: 10/08/2023] [Indexed: 10/13/2023]
Abstract
The Kisspeptin1 (KISS1)/neurokinin B (NKB)/Dynorphin (Dyn) [KNDy] neurons in the hypothalamus regulate the reproduction stage in human beings and rodents. KNDy neurons co-expressed all KISS1, NKB, and Dyn peptides, and hence commonly regarded as KISS1 neurons. KNDy neurons contribute to the "GnRH pulse generator" and are implicated in the regulation of pulsatile GnRH release. The estradiol (E2)-estrogen receptor (ER) interactions over GnRH neurons in the hypothalamus cause nitric oxide (NO) discharge, in addition to presynaptic GABA and glutamate discharge from respective neurons. The released GABA and glutamate facilitate the activity of GnRH neurons via GABAA-R and AMPA/kainate-R. The KISS1 stimulates MAPK/ERK1/2 signaling and cause the release of Ca2+ from intracellular store, which contribute to neuroendocrine function, increase apoptosis and decrease cell proliferation and metastasis. The ageing in women deteriorates KISS1/KISS1R interaction in the hypothalamus which causes lower levels of GnRH. Because examining the human brain is so challenging, decades of clinical research have failed to find the causes of KNDy/GnRH dysfunction. The KISS1/KISS1R interactions in the brain have a neuroprotective effect against Alzheimer's disease (AD). These findings modulate the pathophysiological role of the KNDy/GnRH neural network in polycystic ovarian syndrome (PCOS) associated with ageing and, its protective role in cancer and AD. This review concludes with protecting effect of the steroid-derived acute regulatory enzyme (StAR) against neurotoxicity in the hippocampus, and hypothalamus, and these measures are fundamental for delaying ageing with PCOS. StAR could serve as novel diagnostic marker and therapeutic target for the most prevalent hormone-sensitive breast cancers (BCs).
Collapse
Affiliation(s)
- Siva Prasad Panda
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India.
| | - Adarsh Kesharwani
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | | | - Dsnbk Prasanth
- KVSR Siddhartha College of Pharmaceutical Sciences, Vijayawada, Andhrapradesh, India
| | - Bhaskara Raju Vatchavai
- Sri Vasavi Institute of Pharmaceutical Sciences, Pedatadepalli, Tadepalligudem, Andhrapradesh, India
| | - P V Kamala Kumari
- Vignan Institute of Pharmaceutical Technology, Duvvada, Visakhapatnam, Andhrapradesh, India
| | | | | |
Collapse
|
3
|
Piet R. Circadian and kisspeptin regulation of the preovulatory surge. Peptides 2023; 163:170981. [PMID: 36842628 DOI: 10.1016/j.peptides.2023.170981] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/19/2023] [Accepted: 02/23/2023] [Indexed: 02/28/2023]
Abstract
Fertility in mammals is ultimately controlled by a small population of neurons - the gonadotropin-releasing hormone (GnRH) neurons - located in the ventral forebrain. GnRH neurons control gonadal function through the release of GnRH, which in turn stimulates the secretion of the anterior pituitary gonadotropins luteinizing hormone (LH) and follicle-stimulating hormone (FSH). In spontaneous ovulators, ovarian follicle maturation eventually stimulates, via sex steroid feedback, the mid-cycle surge in GnRH and LH secretion that causes ovulation. The GnRH/LH surge is initiated in many species just before the onset of activity through processes controlled by the central circadian clock, ensuring that the neuroendocrine control of ovulation and sex behavior are coordinated. This review aims to give an overview of anatomical and functional studies that collectively reveal some of the mechanisms through which the central circadian clock regulates GnRH neurons and their afferent circuits to drive the preovulatory surge.
Collapse
Affiliation(s)
- Richard Piet
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, United States.
| |
Collapse
|
4
|
Prashar V, Arora T, Singh R, Sharma A, Parkash J. Hypothalamic Kisspeptin Neurons: Integral Elements of the GnRH System. Reprod Sci 2023; 30:802-822. [PMID: 35799018 DOI: 10.1007/s43032-022-01027-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/23/2022] [Indexed: 12/19/2022]
Abstract
Highly sophisticated and synchronized interactions of various cells and hormonal signals are required to make organisms competent for reproduction. GnRH neurons act as a common pathway for multiple cues for the onset of puberty and attaining reproductive function. GnRH is not directly receptive to most of the signals required for the GnRH secretion during the various phases of the ovarian cycle. Kisspeptin neurons of the hypothalamus convey these signals required for the synchronized release of the GnRH. The steroid-sensitive anteroventral periventricular nucleus (AVPV) kisspeptin and arcuate nucleus (ARC) KNDy neurons convey steroid feedback during the reproductive cycle necessary for GnRH surge and pulse, respectively. AVPV region kisspeptin neurons also communicate with nNOS synthesizing neurons and suprachiasmatic nucleus (SCN) neurons to coordinate the process of the ovarian cycle. Neurokinin B (NKB) and dynorphin play roles in the GnRH pulse stimulation and inhibition, respectively. The loss of NKB and kisspeptin function results in the development of neuroendocrine disorders such as hypogonadotropic hypogonadism (HH) and infertility. Ca2+ signaling is essential for GnRH pulse generation, which is propagated through gap junctions between astrocytes-KNDy and KNDy-KNDy neurons. Impaired functioning of KNDy neurons could develop the characteristics associated with polycystic ovarian syndrome (PCOS) in rodents. Kisspeptin-increased synthesis led to excessive secretion of the LH associated with PCOS. This review provides the latest insights and understanding into the role of the KNDy and AVPV/POA kisspeptin neurons in GnRH secretion and PCOS.
Collapse
Affiliation(s)
- Vikash Prashar
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Tania Arora
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Randeep Singh
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Arti Sharma
- Department of Computational Sciences, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Jyoti Parkash
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India.
| |
Collapse
|
5
|
Jamieson BB, Piet R. Kisspeptin neuron electrophysiology: Intrinsic properties, hormonal modulation, and regulation of homeostatic circuits. Front Neuroendocrinol 2022; 66:101006. [PMID: 35640722 DOI: 10.1016/j.yfrne.2022.101006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 05/05/2022] [Accepted: 05/19/2022] [Indexed: 11/04/2022]
Abstract
The obligatory role of kisspeptin (KISS1) and its receptor (KISS1R) in regulating the hypothalamic-pituitary-gonadal axis, puberty and fertility was uncovered in 2003. In the few years that followed, an impressive body of work undertaken in many species established that neurons producing kisspeptin orchestrate gonadotropin-releasing hormone (GnRH) neuron activity and subsequent GnRH and gonadotropin hormone secretory patterns, through kisspeptin-KISS1R signaling, and mediate many aspects of gonadal steroid hormone feedback regulation of GnRH neurons. Here, we review knowledge accrued over the past decade, mainly in genetically modified mouse models, of the electrophysiological properties of kisspeptin neurons and their regulation by hormonal feedback. We also discuss recent progress in our understanding of the role of these cells within neuronal circuits that control GnRH neuron activity and GnRH secretion, energy balance and, potentially, other homeostatic and reproductive functions.
Collapse
Affiliation(s)
| | - Richard Piet
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, USA.
| |
Collapse
|
6
|
Sheep as a model for neuroendocrinology research. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 189:1-34. [PMID: 35595346 DOI: 10.1016/bs.pmbts.2022.01.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Animal models remain essential to understand the fundamental mechanisms of physiology and pathology. Particularly, the complex and dynamic nature of neuroendocrine cells of the hypothalamus make them difficult to study. The neuroendocrine systems of the hypothalamus are critical for survival and reproduction, and are highly conserved throughout vertebrate evolution. Their roles in controlling body metabolism, growth and body composition, stress, electrolyte balance, and reproduction, have been intensively studied, and have yielded groundbreaking discoveries. Many of these discoveries would not have been feasible without the use of the domestic sheep (Ovis aries). The sheep has been used for decades to study the neuroendocrine systems of the hypothalamus and has become a model for human neuroendocrinology. The aim of this chapter is to review some of the profound biomedical discoveries made possible by the use of sheep. The advantages and limitations of sheep as a neuroendocrine model will be discussed. While no animal model can perfectly recapitulate a human disease or condition, sheep are invaluable for enabling manipulations not possible in human subjects and isolating physiologic variables to garner insight into neuroendocrinology and associated pathologies.
Collapse
|
7
|
Goodman RL, Herbison AE, Lehman MN, Navarro VM. Neuroendocrine control of gonadotropin-releasing hormone: Pulsatile and surge modes of secretion. J Neuroendocrinol 2022; 34:e13094. [PMID: 35107859 PMCID: PMC9948945 DOI: 10.1111/jne.13094] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 11/28/2022]
Abstract
The concept that different systems control episodic and surge secretion of gonadotropin-releasing hormone (GnRH) was well established by the time that GnRH was identified and formed the framework for studies of the physiological roles of GnRH, and later kisspeptin. Here, we focus on recent studies identifying the neural mechanisms underlying these two modes of secretion, with an emphasis on their core components. There is now compelling data that kisspeptin neurons in the arcuate nucleus that also contain neurokinin B (NKB) and dynorphin (i.e., KNDy cells) and their projections to GnRH dendrons constitute the GnRH pulse generator in mice and rats. There is also strong evidence for a similar role for KNDy neurons in sheep and goats, and weaker data in monkeys and humans. However, whether KNDy neurons act on GnRH dendrons and/or GnRH soma and dendrites that are found in the mediobasal hypothalamus (MBH) of these species remains unclear. The core components of the GnRH/luteinising hormone surge consist of an endocrine signal that initiates the process and a neural trigger that drives GnRH secretion during the surge. In all spontaneous ovulators, the core endocrine signal is a rise in estradiol secretion from the maturing follicle(s), with the site of estrogen positive feedback being the rostral periventricular kisspeptin neurons in rodents and neurons in the MBH of sheep and primates. There is considerable species variations in the neural trigger, with three major classes. First, in reflex ovulators, this trigger is initiated by coitus and carried to the hypothalamus by neural or vascular pathways. Second, in rodents, there is a time of day signal that originates in the suprachiasmatic nucleus and activates rostral periventricular kisspeptin neurons and GnRH soma and dendrites. Finally, in sheep nitric oxide-producing neurons in the ventromedial nucleus, KNDy neurons and rostral kisspeptin neurons all appear to participate in driving GnRH release during the surge.
Collapse
Affiliation(s)
- Robert L. Goodman
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV, USA
| | - Allan E. Herbison
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Michael N. Lehman
- Brain Health Research Institute, Kent State University, Kent, OH, USA
- Department of Biological Sciences, Kent State University, Kent, OH, USA
| | - Victor M. Navarro
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School and Department of Medicine, Boston, MA, USA
| |
Collapse
|
8
|
Constantin S, Moenter SM, Piet R. The electrophysiologic properties of gonadotropin-releasing hormone neurons. J Neuroendocrinol 2022; 34:e13073. [PMID: 34939256 PMCID: PMC9163209 DOI: 10.1111/jne.13073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 11/10/2021] [Accepted: 11/18/2021] [Indexed: 11/26/2022]
Abstract
For about two decades, recordings of identified gonadotropin-releasing hormone (GnRH) neurons have provided a wealth of information on their properties. We describe areas of consensus and debate the intrinsic electrophysiologic properties of these cells, their response to fast synaptic and neuromodulatory input, Ca2+ imaging correlates of action potential firing, and signaling pathways regulating these aspects. How steroid feedback and development change these properties, functions of GnRH neuron subcompartments and local networks, as revealed by chemo- and optogenetic approaches, are also considered.
Collapse
Affiliation(s)
- Stephanie Constantin
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892-3703, USA
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | - Suzanne M Moenter
- Departments of Molecular & Integrative Physiology, Internal Medicine, Obstetrics & Gynecology, and the Reproductive Sciences Program, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Richard Piet
- Brain Health Research Institute & Department of Biological Sciences, Kent State University, Kent, OH, 44242, USA
| |
Collapse
|
9
|
Raúl Schmidt A, Ignacio Felipe Inserra P, Andrés Cortasa S, Proietto S, Fidel V, Halperin J, Daniel Vitullo A, Berta Dorfman V. Distribution of kisspeptin system and its relation with gonadotropin-releasing hormone in the hypothalamus of the South American plains vizcacha, Lagostomus maximus. Gen Comp Endocrinol 2022; 317:113974. [PMID: 34973969 DOI: 10.1016/j.ygcen.2021.113974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 12/21/2021] [Accepted: 12/26/2021] [Indexed: 11/20/2022]
Abstract
Kisspeptin (KISS), a key hormone involved in the regulation of the hypothalamic-pituitary-ovarian (HPO) axis, has been localized in the anteroventral periventricular (AVPV) nucleus and the neighboring rostral periventricular nucleus (PeVN), and in the arcuate (ARC) nucleus of the mammalian hypothalamus. In the ARC, the KISS neurons that co-express neurokinin B (NKB) and dynorphin A (Dyn) are named KNDy cells. The South American plains vizcacha is a rodent with peculiar reproductive traits. Around mid-pregnancy, vizcacha shows the reactivation of its HPO axis with the pulsatile release of gonadotropin-releasing hormone (GnRH) and luteinizing hormone (LH), an essential event for the success of gestation. Considering the role of KISS system in GnRH modulation, the aim of this work was to study their neuroanatomical distribution in adult vizcachas. AVPV showed sexual dimorphism with a significant smaller area in males (t-Test, p < 0.05), and KISS immunoreactivity was detected in somas and varicosities homogenously distributed in the AVPV with a concordant sex-related expression pattern. NKB and Dyn expression was also observed in cytoplasm of neurons scattered in the AVPV. Three subpopulations of neurons were detected in the AVPV: neurons expressing Dyn and NKB (DyNK cells), neurons expressing KISS and NKB (KiNK cells), and single NKB expressing neurons. Strikingly, KISS and Dyn were always expressed in different cells. In addition, in the ARC nucleus, KNDy cells were detected. On the other hand, KISS and GnRH expression was detected in different subpopulations of neurons, GnRH cells showed KISS receptor (KISSR or GPR-54) expression, and KISS immunoreactive afferent contacts were detected making close appositions onto somas and dendrites of GnRH cells. These results show similarities and differences between the KISS system in the hypothalamus of the vizcacha and other mammals, and constitute crucial observations about KISS and GnRH relation. Considering the peculiarity of HPO axis regulation in this species, the present work provides a neuroanatomical framework for the further elucidation of molecular mechanisms underlying GnRH expression and secretion.
Collapse
Affiliation(s)
- Alejandro Raúl Schmidt
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Pablo Ignacio Felipe Inserra
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Santiago Andrés Cortasa
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Sofía Proietto
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Victoria Fidel
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina
| | - Julia Halperin
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Alfredo Daniel Vitullo
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Verónica Berta Dorfman
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina.
| |
Collapse
|
10
|
Ikegami K, Watanabe Y, Nakamura S, Goto T, Inoue N, Uenoyama Y, Tsukamura H. Cellular and molecular mechanisms regulating the KNDy neuronal activities to generate and modulate GnRH pulse in mammals. Front Neuroendocrinol 2022; 64:100968. [PMID: 34808231 DOI: 10.1016/j.yfrne.2021.100968] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/18/2021] [Accepted: 11/15/2021] [Indexed: 12/30/2022]
Abstract
Accumulating findings during the past decades have demonstrated that the hypothalamic arcuate kisspeptin neurons are supposed to be responsible for pulsatile release of gonadotropin-releasing hormone (GnRH) to regulate gametogenesis and steroidogenesis in mammals. The arcuate kisspeptin neurons express neurokinin B (NKB) and dynorphin A (Dyn), thus, the neurons are also referred to as KNDy neurons. In the present article, we mainly focus on the cellular and molecular mechanisms underlying GnRH pulse generation, that is focused on the action of NKB and Dyn and an interaction between KNDy neurons and astrocytes to control GnRH pulse generation. Then, we also discuss the factors that modulate the activity of KNDy neurons and consequent pulsatile GnRH/LH release in mammals.
Collapse
Affiliation(s)
- Kana Ikegami
- Graduate School of Science, The University of Tokyo, Tokyo 113-0033, Japan
| | - Youki Watanabe
- Graduate School of Applied Life Science, Nippon Veterinary and Life Science University, Tokyo 180-8602, Japan
| | - Sho Nakamura
- Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Ehime 794-8555, Japan
| | - Teppei Goto
- RIKEN Center for Biosystems Dynamics Research, Hyogo 650-0047, Japan
| | - Naoko Inoue
- Graduate School of Bioagricultural Science, Nagoya University, Nagoya 464-8601, Japan
| | - Yoshihisa Uenoyama
- Graduate School of Bioagricultural Science, Nagoya University, Nagoya 464-8601, Japan
| | - Hiroko Tsukamura
- Graduate School of Bioagricultural Science, Nagoya University, Nagoya 464-8601, Japan.
| |
Collapse
|
11
|
Herbison AE. The dendron and episodic neuropeptide release. J Neuroendocrinol 2021; 33:e13024. [PMID: 34427000 DOI: 10.1111/jne.13024] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/04/2021] [Accepted: 08/05/2021] [Indexed: 12/21/2022]
Abstract
The unexpected observation that the long processes of gonadotrophin-releasing hormone (GnRH) neurons not only conducted action potentials, but also operated to integrate afferent information at their distal-most extent gave rise to the concept of a blended dendritic-axonal process termed the "dendron". The proximal dendrites of the GnRH neuron function in a conventional manner, receiving synaptic inputs and initiating action potentials that are critical for the surge mode of GnRH secretion. The distal dendrons are regulated by both classical synapses and volume transmission and likely operate using subthreshold electrotonic propagation into the nearby axon terminals in the median eminence. Evidence indicates that neural processing at the distal dendron is responsible for the pulsatile patterning of GnRH secretion. Although the dendron remains unique to the GnRH neuron, data show that it exists in both mice and rats and may be a common feature of mammalian species in which GnRH neuron cell bodies do not migrate into the basal hypothalamus. This review outlines the discovery and function of the dendron as a unique neuronal structure optimised to generate episodic neuronal output.
Collapse
Affiliation(s)
- Allan E Herbison
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, UK
| |
Collapse
|
12
|
Arcuate and Preoptic Kisspeptin Neurons Exhibit Differential Projections to Hypothalamic Nuclei and Exert Opposite Postsynaptic Effects on Hypothalamic Paraventricular and Dorsomedial Nuclei in the Female Mouse. eNeuro 2021; 8:ENEURO.0093-21.2021. [PMID: 34281980 PMCID: PMC8354717 DOI: 10.1523/eneuro.0093-21.2021] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 06/21/2021] [Accepted: 07/11/2021] [Indexed: 01/24/2023] Open
Abstract
Kisspeptin (Kiss1) neurons provide indispensable excitatory input to gonadotropin-releasing hormone (GnRH) neurons, which is important for the coordinated release of gonadotropins, estrous cyclicity and ovulation. However, Kiss1 neurons also send projections to many other brain regions within and outside the hypothalamus. Two different populations of Kiss1 neurons, one in the arcuate nucleus (Kiss1ARH) and another in the anteroventral periventricular nucleus (AVPV) and periventricular nucleus (PeN; Kiss1AVPV/PeN) of the hypothalamus are differentially regulated by ovarian steroids, and are believed to form direct contacts with GnRH neurons as well as other neurons. To investigate the projection fields from Kiss1AVPV/PeN and Kiss1ARH neurons in female mice, we used anterograde projection analysis, and channelrhodopsin-assisted circuit mapping (CRACM) to explore their functional input to select target neurons within the paraventricular (PVH) and dorsomedial (DMH) hypothalamus, key preautonomic nuclei. Cre-dependent viral (AAV1-DIO-ChR2 mCherry) vectors were injected into the brain to label the two Kiss1 neuronal populations. Immunocytochemistry (ICC) for mCherry and neuropeptides combined with confocal microscopy was used to determine the projection-fields of both Kiss1 neuronal groups. Whole-cell electrophysiology and optogenetics were used to elucidate the functional input to the PVH and DMH. Our analysis revealed many common but also several clearly separate projection fields between the two different populations of Kiss1 neurons. In addition, optogenetic stimulation of Kiss1 projections to PVH prodynorphin, Vglut2 and DMH CART-expressing neurons, revealed excitatory glutamatergic input from Kiss1ARH neurons and inhibitory GABAergic input from Kiss1AVPV/PeN neurons. Therefore, these steroid-sensitive Kiss1 neuronal groups can differentially control the excitability of target neurons to coordinate autonomic functions with reproduction.
Collapse
|
13
|
Berland M, Paiva L, Santander LA, Ratto MH. Distribution of GnRH and Kisspeptin Immunoreactivity in the Female Llama Hypothalamus. Front Vet Sci 2021; 7:597921. [PMID: 33604362 PMCID: PMC7884347 DOI: 10.3389/fvets.2020.597921] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 11/30/2020] [Indexed: 12/05/2022] Open
Abstract
Llamas are induced non-reflex ovulators, which ovulate in response to the hormonal stimulus of the male protein beta-nerve growth factor (β-NGF) that is present in the seminal plasma; this response is dependent on the preovulatory gonadotrophin-releasing hormone (GnRH) release from the hypothalamus. GnRH neurones are vital for reproduction, as these provide the input that controls the release of luteinizing hormone (LH) and follicle-stimulating hormone (FSH) from the pituitary gland. However, in spontaneous ovulators, the activity of GnRH cells is regulated by kisspeptin neurones that relay the oestrogen signal arising from the periphery. Here, we investigated the organisation of GnRH and kisspeptin systems in the hypothalamus of receptive adult female llamas. We found that GnRH cells exhibiting different shapes were distributed throughout the ventral forebrain and some of these were located in proximity to blood vessels; sections of the mediobasal hypothalamus (MBH) displayed the highest number of cells. GnRH fibres were observed in both the organum vasculosum laminae terminalis (OVLT) and median eminence (ME). We also detected abundant kisspeptin fibres in the MBH and ME; kisspeptin cells were found in the arcuate nucleus (ARC), but not in rostral areas of the hypothalamus. Quantitative analysis of GnRH and kisspeptin fibres in the ME revealed a higher innervation density of kisspeptin than of GnRH fibres. The physiological significance of the anatomical findings reported here for the ovulatory mechanism in llamas is still to be determined.
Collapse
Affiliation(s)
- Marco Berland
- Departamento de Ciencias Veterinarias y Salud Pública, Facultad de Recursos Naturales, Universidad Católica de Temuco, Temuco, Chile
| | - Luis Paiva
- Instituto de Ciencia Animal, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Lig Alondra Santander
- Departamento de Ciencias Veterinarias y Salud Pública, Facultad de Recursos Naturales, Universidad Católica de Temuco, Temuco, Chile
| | - Marcelo Héctor Ratto
- Instituto de Ciencia Animal, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| |
Collapse
|
14
|
Liu X, Yeo SH, McQuillan HJ, Herde MK, Hessler S, Cheong I, Porteous R, Herbison AE. Highly redundant neuropeptide volume co-transmission underlying episodic activation of the GnRH neuron dendron. eLife 2021; 10:62455. [PMID: 33464205 PMCID: PMC7847305 DOI: 10.7554/elife.62455] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 01/15/2021] [Indexed: 12/16/2022] Open
Abstract
The necessity and functional significance of neurotransmitter co-transmission remains unclear. The glutamatergic 'KNDy' neurons co-express kisspeptin, neurokinin B (NKB), and dynorphin and exhibit a highly stereotyped synchronized behavior that reads out to the gonadotropin-releasing hormone (GnRH) neuron dendrons to drive episodic hormone secretion. Using expansion microscopy, we show that KNDy neurons make abundant close, non-synaptic appositions with the GnRH neuron dendron. Electrophysiology and confocal GCaMP6 imaging demonstrated that, despite all three neuropeptides being released from KNDy terminals, only kisspeptin was able to activate the GnRH neuron dendron. Mice with a selective deletion of kisspeptin from KNDy neurons failed to exhibit pulsatile hormone secretion but maintained synchronized episodic KNDy neuron behavior that is thought to depend on recurrent NKB and dynorphin transmission. This indicates that KNDy neurons drive episodic hormone secretion through highly redundant neuropeptide co-transmission orchestrated by differential post-synaptic neuropeptide receptor expression at the GnRH neuron dendron and KNDy neuron.
Collapse
Affiliation(s)
- Xinhuai Liu
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Shel-Hwa Yeo
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand.,Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - H James McQuillan
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Michel K Herde
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Sabine Hessler
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Isaiah Cheong
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Robert Porteous
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Allan E Herbison
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand.,Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
15
|
Uenoyama Y, Nagae M, Tsuchida H, Inoue N, Tsukamura H. Role of KNDy Neurons Expressing Kisspeptin, Neurokinin B, and Dynorphin A as a GnRH Pulse Generator Controlling Mammalian Reproduction. Front Endocrinol (Lausanne) 2021; 12:724632. [PMID: 34566891 PMCID: PMC8458932 DOI: 10.3389/fendo.2021.724632] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 08/23/2021] [Indexed: 01/16/2023] Open
Abstract
Increasing evidence accumulated during the past two decades has demonstrated that the then-novel kisspeptin, which was discovered in 2001, the known neuropeptides neurokinin B and dynorphin A, which were discovered in 1983 and 1979, respectively, and their G-protein-coupled receptors, serve as key molecules that control reproduction in mammals. The present review provides a brief historical background and a summary of our recent understanding of the roles of hypothalamic neurons expressing kisspeptin, neurokinin B, and dynorphin A, referred to as KNDy neurons, in the central mechanism underlying gonadotropin-releasing hormone (GnRH) pulse generation and subsequent tonic gonadotropin release that controls mammalian reproduction.
Collapse
|
16
|
Lin XH, Lass G, Kong LS, Wang H, Li XF, Huang HF, O’Byrne KT. Optogenetic Activation of Arcuate Kisspeptin Neurons Generates a Luteinizing Hormone Surge-Like Secretion in an Estradiol-Dependent Manner. Front Endocrinol (Lausanne) 2021; 12:775233. [PMID: 34795643 PMCID: PMC8593229 DOI: 10.3389/fendo.2021.775233] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 10/18/2021] [Indexed: 01/31/2023] Open
Abstract
Traditionally, the anteroventral periventricular (AVPV) nucleus has been the brain area associated with luteinizing hormone (LH) surge secretion in rodents. However, the role of the other population of hypothalamic kisspeptin neurons, in the arcuate nucleus (ARC), has been less well characterized with respect to surge generation. Previous experiments have demonstrated ARC kisspeptin knockdown reduced the amplitude of LH surges, indicating that they have a role in surge amplification. The present study used an optogenetic approach to selectively stimulate ARC kisspeptin neurons and examine the effect on LH surges in mice with different hormonal administrations. LH level was monitored from 13:00 to 21:00 h, at 30-minute intervals. Intact Kiss-Cre female mice showed increased LH secretion during the stimulation period in addition to displaying a spontaneous LH surge around the time of lights off. In ovariectomized Kiss-Cre mice, optogenetic stimulation was followed by a surge-like secretion of LH immediately after the stimulation period. Ovariectomized Kiss-Cre mice with a low dose of 17β-estradiol (OVX+E) replacement displayed a surge-like increase in LH release during period of optic stimulation. No LH response to the optic stimulation was observed in OVX+E mice on the day of estradiol benzoate (EB) treatment (day 1). However, after administration of progesterone (day 2), all OVX+E+EB+P mice exhibited an LH surge during optic stimulation. A spontaneous LH surge also occurred in these mice at the expected time. Taken together, these results help to affirm the fact that ARC kisspeptin may have a novel amplificatory role in LH surge production, which is dependent on the gonadal steroid milieu.
Collapse
Affiliation(s)
- Xian-Hua Lin
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
- Department of Women and Children’s Health, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Geffen Lass
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Ling-Si Kong
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Hui Wang
- Department of Obstetrics and Gynecology, Maternal and Child Health Hospital of Songjiang District, Shanghai, China
| | - Xiao-Feng Li
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - He-Feng Huang
- Department of Women and Children’s Health, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Kevin T. O’Byrne
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
- *Correspondence: Kevin T. O’Byrne, kevin.o’
| |
Collapse
|
17
|
Yip SH, Campos P, Liu X, Porteous R, Herbison AE. Innervation of GnRH Neuron Distal Projections and Activation by Kisspeptin in a New GnRH-Cre Rat Model. Endocrinology 2021; 162:bqaa186. [PMID: 33057587 DOI: 10.1210/endocr/bqaa186] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Indexed: 12/12/2022]
Abstract
The neural mechanisms generating pulsatile GnRH release from the median eminence (ME) remain unclear. Studies undertaken in the mouse demonstrate that GnRH neurons extend projections to the ME that have properties of both dendrites and axons, termed "dendrons," and that the kisspeptin neuron pulse generator targets these distal dendrons to drive pulsatile GnRH secretion. It presently remains unknown whether the GnRH neuron dendron exists in other species. We report here the generation of a knock-in Gnrh1-Ires-Cre rat line with near-perfect targeting of Cre recombinase to the GnRH neuronal phenotype. More than 90% of adult male and female GnRH neurons express Cre with no ectopic expression. Adeno-associated viruses were used in adult female Gnrh1-Ires-Cre rats to target mCherry or GCAMP6 to rostral preoptic area GnRH neurons. The mCherry tracer revealed the known unipolar and bipolar morphology of GnRH neurons and their principal projection pathways to the external zone of the ME. Synaptophysin-labeling of presynaptic nerve terminals revealed that GnRH neuron distal projections received numerous close appositions as they passed through the arcuate nucleus and into the median eminence. Confocal GCaMP6 imaging in acute horizontal brain slices demonstrated that GnRH neuron distal projections lateral to the median eminence were activated by kisspeptin. These studies indicate the presence of a dendron-like arrangement in the rat with GnRH neuron distal projections receiving synaptic input and responding to kisspeptin.
Collapse
Affiliation(s)
- Siew Hoong Yip
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Pauline Campos
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Xinhuai Liu
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Robert Porteous
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Allan E Herbison
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| |
Collapse
|
18
|
Patel R, Smith JT. Novel actions of kisspeptin signaling outside of GnRH-mediated fertility: a potential role in energy balance. Domest Anim Endocrinol 2020; 73:106467. [PMID: 32278499 DOI: 10.1016/j.domaniend.2020.106467] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 02/24/2020] [Accepted: 02/26/2020] [Indexed: 01/08/2023]
Abstract
Kisspeptin, encoded by Kiss1 gene expressing neurons in the hypothalamus, is a requisite for fertility and now appears critical in the regulation of energy balance. Kisspeptin neurons, particularly those in the arcuate nucleus (ARC), receive information directly and indirectly from a diverse array of brain regions including the bed nucleus of the stria terminalis, amygdala, interpeduncular nucleus, hippocampus, and cortex. On the other hand, kisspeptin neuron projections clearly extend to GnRH neuron cell bodies in rodents, sheep, and primates and beyond to other-non-GnRH-brain areas. Kiss1r, the kisspeptin receptor, is expressed on GnRH neurons and also in additional brain areas and peripheral tissues, indicating a nonreproductive role. Kisspeptin neurons clearly receive signals pertinent to deviations in energy balance but are now recognized as a novel neuroendocrine player in the fine balance of energy intake and expenditure. Mice that have a dysfunctional gene for Kiss1r develop an obese and diabetic phenotype. The mechanism behind this altered metabolic state is still mostly unknown; however, Kiss1r expression in the pancreas and brown adipose tissue is clearly functional and required for normal glucose tolerance and energy expenditure, respectively. Kisspeptin neurons in the ARC also participate in the generation of circadian rhythms, specifically those concerning food intake and metabolism, offering a potential explanation for the obesity in Kiss1r knockout mice. Overall, the discoveries of new mechanistic roles for kisspeptin in both normal and pathophysiologic states of energy balance may lead to further understating of obesity prevalence and novel therapeutic targets and interventions.
Collapse
Affiliation(s)
- R Patel
- School of Human Sciences, M309, The University of Western Australia, 35 Stirling Highway Crawley, Perth, Western Australia, Australia 6009
| | - J T Smith
- School of Human Sciences, M309, The University of Western Australia, 35 Stirling Highway Crawley, Perth, Western Australia, Australia 6009.
| |
Collapse
|
19
|
Etzion T, Zmora N, Zohar Y, Levavi-Sivan B, Golan M, Gothilf Y. Ectopic over expression of kiss1 may compensate for the loss of kiss2. Gen Comp Endocrinol 2020; 295:113523. [PMID: 32470472 DOI: 10.1016/j.ygcen.2020.113523] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 05/19/2020] [Accepted: 05/22/2020] [Indexed: 10/24/2022]
Abstract
Kisspeptin (KISS) is a neuropeptide which plays a central role in the regulation of the hypothalamic-pituitary-gonadal axis, and is essential for sexual maturation and fertility in mammals. Unlike mammals, which possess only one KISS gene, two paralogous genes, kiss1 and kiss2, have been identified in zebrafish and other non-mammalian vertebrates. Previous studies suggest that Kiss2, but not Kiss1, is the reproduction relevant form amongst the two. To better understand the role of each of these isoforms in reproduction, a loss of function approach was applied. Two genetic manipulation techniques-clustered regularly interspaced short palindromic repeats (CRISPR) and transcription activator-like effector nucleases (TALEN)-were used to generate kiss1 and kiss2 knockout (KO) zebrafish lines, respectively. Examination of these KO lines showed that reproductive capability was not impaired, confirming earlier observations. Further analysis revealed that KO of kiss2 caused a significant increase in expression levels of kiss1, kiss2r and tac3a, while KO of kiss1 had no effect on the expression of any of the examined genes. In situ hybridization analysis revealed that kiss1 mRNA is expressed only in the habenula in wild type brains, while in kiss2 KO fish, kiss1 mRNA-expressing cells were identified also in the ventral telencephalon, the ventral part of the entopeduncular nucleus, and the dorsal and ventral hypothalamus. Interestingly, these regions are known to express kiss2r, and the ventral hypothalamus normally expresses kiss2. These results suggest that a compensatory mechanism, involving ectopic kiss1 expression, takes place in the kiss2 KO fish, which may substitute for Kiss2 activity.
Collapse
Affiliation(s)
- Talya Etzion
- Department of Neurobiology, George S. Wise Faculty of Life Sciences and Sagol School of Neurosciences, University of Tel Aviv, Tel Aviv 69978, Israel
| | - Nilli Zmora
- Department of Marine Biotechnology, Institute of Marine and Environmental Technology, University of Maryland Baltimore County, Baltimore, MD, USA
| | - Yonatan Zohar
- Department of Marine Biotechnology, Institute of Marine and Environmental Technology, University of Maryland Baltimore County, Baltimore, MD, USA
| | - Berta Levavi-Sivan
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - Matan Golan
- Department of Poultry and Aquaculture, Institute of Animal Sciences, Agricultural Research Organization, Volcani Center, Rishon Letziyon 7505101, Israel
| | - Yoav Gothilf
- Department of Neurobiology, George S. Wise Faculty of Life Sciences and Sagol School of Neurosciences, University of Tel Aviv, Tel Aviv 69978, Israel.
| |
Collapse
|
20
|
Lass G, Li XF, de Burgh RA, He W, Kang Y, Hwa-Yeo S, Sinnett-Smith LC, Manchishi SM, Colledge WH, Lightman SL, O'Byrne KT. Optogenetic stimulation of kisspeptin neurones within the posterodorsal medial amygdala increases luteinising hormone pulse frequency in female mice. J Neuroendocrinol 2020; 32:e12823. [PMID: 31872920 PMCID: PMC7116078 DOI: 10.1111/jne.12823] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 11/25/2019] [Accepted: 12/19/2019] [Indexed: 12/11/2022]
Abstract
Kisspeptin within the arcuate nucleus of the hypothalamus is a critical neuropeptide in the regulation of reproduction. Together with neurokinin B and dynorphin A, arcuate kisspeptin provides the oscillatory activity that drives the pulsatile secretion of gonadotrophin-releasing hormone (GnRH), and therefore luteinising hormone (LH) pulses, and is considered to be a central component of the GnRH pulse generator. It is well established that the amygdala also exerts an influence over gonadotrophic hormone secretion and reproductive physiology. The discovery of kisspeptin and its receptor within the posterodorsal medial amygdala (MePD) and our recent finding showing that intra-MePD administration of kisspeptin or a kisspeptin receptor antagonist results in increased LH secretion and decreased LH pulse frequency, respectively, suggests an important role for amygdala kisspeptin signalling in the regulation of the GnRH pulse generator. To further investigate the function of amygdala kisspeptin, the present study used an optogenetic approach to selectively stimulate MePD kisspeptin neurones and examine the effect on pulsatile LH secretion. MePD kisspeptin neurones in conscious Kiss1-Cre mice were virally infected to express the channelrhodopsin 2 protein and selectively stimulated by light via a chronically implanted fibre optic cannula. Continuous stimulation using 5 Hz resulted in an increased LH pulse frequency, which was not observed at the lower stimulation frequencies of 0.5 and 2 Hz. In wild-type animals, continuous stimulation at 5 Hz did not affect LH pulse frequency. These results demonstrate that selective activation of MePD Kiss1 neurones can modulate hypothalamic GnRH pulse generator frequency.
Collapse
Affiliation(s)
- Geffen Lass
- Department of Women and Children's Health, Faculty of Life Sciences and Medicine, King's College London Guy's Campus, London, UK
| | - Xiao Feng Li
- Department of Women and Children's Health, Faculty of Life Sciences and Medicine, King's College London Guy's Campus, London, UK
| | - Ross A de Burgh
- Department of Women and Children's Health, Faculty of Life Sciences and Medicine, King's College London Guy's Campus, London, UK
| | - Wen He
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yanping Kang
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Shel Hwa-Yeo
- Reproductive Physiology Group, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Lydia C Sinnett-Smith
- Reproductive Physiology Group, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Stephen M Manchishi
- Reproductive Physiology Group, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - William H Colledge
- Reproductive Physiology Group, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Stafford Louis Lightman
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, The Dorothy Hodgkin Building, University of Bristol, Bristol, UK
| | - Kevin T O'Byrne
- Department of Women and Children's Health, Faculty of Life Sciences and Medicine, King's College London Guy's Campus, London, UK
| |
Collapse
|
21
|
Abstract
This article contains a systematic review of the main developments that have occurred in the area of male hypogonadism between the publication of the Endocrine Society Guidelines of 2010 and 2018 and after 2018.
Collapse
Affiliation(s)
- Marco Marcelli
- Department of Medicine, Division of Endocrinology, Baylor College of Medicine, Houson, Texas, USA .,Section of Endocrinology, Michael E DeBakey VA Medical Center, Houston, Texas, USA
| | - Sanjay Navin Mediwala
- Department of Medicine, Division of Endocrinology, Baylor College of Medicine, Houson, Texas, USA.,Section of Endocrinology, Michael E DeBakey VA Medical Center, Houston, Texas, USA
| |
Collapse
|
22
|
Gotlieb N, Baker CN, Moeller J, Kriegsfeld LJ. Time-of-day-dependent sensitivity of the reproductive axis to RFamide-related peptide-3 inhibition in female Syrian hamsters. J Neuroendocrinol 2019; 31:e12798. [PMID: 31550401 PMCID: PMC6991702 DOI: 10.1111/jne.12798] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 08/15/2019] [Accepted: 09/19/2019] [Indexed: 12/15/2022]
Abstract
In spontaneously ovulating rodent species, the timing of the luteinising hormone (LH) surge is controlled by the master circadian pacemaker in the suprachiasmatic nucleus (SCN). The SCN initiates the LH surge via the coordinated control of two opposing neuropeptidergic systems that lie upstream of the gonadotrophin-releasing hormone (GnRH) neuronal system: the stimulatory peptide, kisspeptin, and the inhibitory peptide, RFamide-related peptide-3 (RFRP-3; the mammalian orthologue of avian gonadotrophin-inhibitory hormone [GnIH]). We have previously shown that the GnRH system exhibits time-dependent sensitivity to kisspeptin stimulation, further contributing to the precise timing of the LH surge. To examine whether this time-dependent sensitivity of the GnRH system is unique to kisspeptin or a more common mechanism of regulatory control, we explored daily changes in the response of the GnRH system to RFRP-3 inhibition. Female Syrian hamsters were ovariectomised to eliminate oestradiol (E2 )-negative-feedback and RFRP-3 or saline was centrally administered in the morning or late afternoon. LH concentrations and Lhβ mRNA expression did not differ between morning RFRP-3-and saline-treated groups, although they were markedly suppressed by RFRP-3 administration in the afternoon. However, RFRP-3 inhibition of circulating LH at the time of the surge does not appear to act via the GnRH system because no differences in medial preoptic area Gnrh or RFRP-3 receptor Gpr147 mRNA expression were observed. Rather, RFRP-3 suppressed arcuate nucleus Kiss1 mRNA expression and potentially impacted pituitary gonadotrophs directly. Taken together, these findings reveal time-dependent responsiveness of the reproductive axis to RFRP-3 inhibition, possibly via variation in the sensitivity of arcuate nucleus kisspeptin neurones to this neuropeptide.
Collapse
Affiliation(s)
- Neta Gotlieb
- Department of Psychology, University of California Berkeley, Berkeley, CA, USA
| | - Cydni N. Baker
- Department of Psychology, University of California Berkeley, Berkeley, CA, USA
| | - Jacob Moeller
- Department of Integrative Biology, University of California Berkeley, Berkeley, CA, USA
| | - Lance J. Kriegsfeld
- Department of Psychology, University of California Berkeley, Berkeley, CA, USA
- Department of Integrative Biology, University of California Berkeley, Berkeley, CA, USA
- Graduate Group in Endocrinology, University of California Berkeley, Berkeley, CA, USA
- The Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
| |
Collapse
|
23
|
Polkowska J, Wójcik-G Adysz A, Chmielewska N, Wa Kowska M. Expression of kisspeptin protein in hypothalamus and LH profile of growing female lambs. Reprod Fertil Dev 2019; 30:609-618. [PMID: 28917264 DOI: 10.1071/rd17018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 08/25/2017] [Indexed: 11/23/2022] Open
Abstract
Kisspeptin (kp) is considered to be one of the major regulators of the induction of pubertal events via the activation of the gonadotrophin-releasing hormone-LH system. The aim of the present study was to analyse expression of immunoreactive (ir) kp in the hypothalamic neurons of female lambs from the neonatal to the peripubertal period (5 days to 32 weeks) in relation to the plasma LH pattern using immunohistochemistry and image analysis. Hypothalami were collected from female lambs (n=33) from the infantile, juvenile, prepubertal and peripubertal periods. The population of kp-ir perikarya was detected mainly in the arcuate nucleus and their number increased gradually from 5 to 16 weeks of age and was maintained at a high level up to the peripubertal stage. This was reflected by the significant (P<0.05) gradual increase in the percentage of hypothalamic area occupied by kp-ir neurons and increase in the number of kp-ir perikarya within the arcuate nucleus. The same pattern of kp immunoreactivity was observed in the median eminence. Plasma LH concentration increased from Week 5 to Weeks 12-16 and further increased at Week 32. LH pulse frequency increased from Week 5 to 32 (P<0.05). Thus, changes in kp expression reflected changes in the LH pattern during lamb growth. The data obtained provide evidence about the participation of kp in the mechanisms of ontogenic development of ovine reproductive processes.
Collapse
Affiliation(s)
- Jolanta Polkowska
- Department of Physiology, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3, 05-110 Jab?onna, Poland
| | - Anna Wójcik-G Adysz
- Department of Physiology, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3, 05-110 Jab?onna, Poland
| | - Natalia Chmielewska
- Department of Neurochemistry, Institute of Psychiatry and Neurology, Sobieskiego 9, 02-957 Warsaw, Poland
| | - Marta Wa Kowska
- Department of Physiology, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3, 05-110 Jab?onna, Poland
| |
Collapse
|
24
|
Costa HC, Da-Silva JM, Diniz GB, Motta-Teixeira LC, Da-Silva RJ, Battagello DS, Sita LV, de-Moraes Machado C, Horta-Júnior JAC, Bittencourt JC. Characterisation and origins of melanin-concentrating hormone immunoreactive fibres of the posterior lobe of the pituitary and median eminence during lactation in the Long-Evans rat. J Neuroendocrinol 2019; 31:e12723. [PMID: 31034718 DOI: 10.1111/jne.12723] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 04/24/2019] [Accepted: 04/25/2019] [Indexed: 11/30/2022]
Abstract
Although the melanin-concentrating hormone (MCH) and its coding mRNA are predominantly found in the tuberal hypothalamus, there is detectable synthesis of MCH in the preoptic hypothalamus exclusively in lactating dams, suggesting a participation of MCH in the alterations that take place after parturition. Also implicated in the dam physiology is oxytocin, a neurohormone released from the posterior pituitary that is necessary for milk ejection. Because the projection fields from oxytocin-immunoreactive (-IR) neurones and the mediobasal preoptic hypothalamus overlap and MCH-IR neurones are found in proximity to oxytocin neurones, we investigated the spatial relationship between MCH and oxytocin fibres. Accordingly, we employed multiple immunohistochemistry labelling for MCH and oxytocin for light and electron microscopy techniques, in addition to i.v. tracer injection combined with in situ hybridisation to identify MCH neurones that project to neurosecretory areas. As described for other strains, lactating Long-Evans dams also display immunoreactivity for MCH in the preoptic hypothalamus on days 12 and 19 of lactation. The appearance of these neurones is contemporaneous with an increase in MCH-IR fibres in both the internal layer of the median eminence and the posterior pituitary. In both regions, MCH- and oxytocin-IR fibres were found in great proximity, although there was no evidence for synaptic interaction between these two populations at the ultrastructural level. The tracer injection revealed that only mediobasal preoptic MCH neurones project to the posterior pituitary, suggesting a neuroendocrine-modulatory role for this population. When taken together, the results obtained in the present study indicate that neuroplasticity events at the mediobasal preoptic hypothalamus that occur during late lactation may be part of a neuroendocrinology control loop involving both MCH and oxytocin.
Collapse
Affiliation(s)
- Helder C Costa
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Joelcimar M Da-Silva
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Giovanne B Diniz
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Livia C Motta-Teixeira
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Renata J Da-Silva
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Daniella S Battagello
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
- Center for Neuroscience and Behavior, Institute of Psychology, University of São Paulo (USP), São Paulo, Brazil
| | - Luciane V Sita
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Carla de-Moraes Machado
- Department of Anatomy, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, Brazil
| | - José A C Horta-Júnior
- Department of Anatomy, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, Brazil
- Electron Microscopy Center, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, Brazil
| | - Jackson C Bittencourt
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
- Center for Neuroscience and Behavior, Institute of Psychology, University of São Paulo (USP), São Paulo, Brazil
| |
Collapse
|
25
|
Uenoyama Y, Inoue N, Nakamura S, Tsukamura H. Central Mechanism Controlling Pubertal Onset in Mammals: A Triggering Role of Kisspeptin. Front Endocrinol (Lausanne) 2019; 10:312. [PMID: 31164866 PMCID: PMC6536648 DOI: 10.3389/fendo.2019.00312] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 04/30/2019] [Indexed: 01/29/2023] Open
Abstract
Pubertal onset is thought to be timed by an increase in pulsatile gonadotropin-releasing hormone (GnRH)/gonadotropin secretion in mammals. The underlying mechanism of pubertal onset in mammals is still an open question. Evidence accumulated in the last 15 years suggests that kisspeptin/neurokinin B/dynorphin A (KNDy) neurons in the hypothalamic arcuate nucleus play a key role in pubertal onset by triggering pulsatile GnRH/gonadotropin secretin in mammals. Specifically, KNDy neurons are now considered a part of GnRH pulse generator, in which neurokinin B facilitates and dynorphin A inhibits, the synchronized discharge of KNDy neurons in autocrine and/or paracrine manners. Kisspeptin serves as a potent secretagogue of GnRH secretion and thus its release is fundamental to pubertal increase in GnRH/gonadotropin secretion in mammals. Proposed mechanisms inhibiting Kiss1 (kisspeptin gene) expression during childhood to juvenile varies from species to species: we envisage that negative feedback action of estrogen plays a key role in the inhibition of Kiss1 expression in KNDy neurons in rodents and sheep, whereas estrogen-independent inhibition of kisspeptin secretion by γ-amino butyric acid or neuropeptide Y are suggested to be responsible for the pre-pubertal suppression of GnRH/gonadotropin secretion in primates. Taken together, the timing of pubertal onset is postulated to be controlled by upstream regulators for kisspeptin biosynthesis and secretion in mammals.
Collapse
Affiliation(s)
- Yoshihisa Uenoyama
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
- *Correspondence: Yoshihisa Uenoyama
| | - Naoko Inoue
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Sho Nakamura
- Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Japan
| | - Hiroko Tsukamura
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| |
Collapse
|
26
|
Uenoyama Y, Inoue N, Maeda KI, Tsukamura H. The roles of kisspeptin in the mechanism underlying reproductive functions in mammals. J Reprod Dev 2018; 64:469-476. [PMID: 30298825 PMCID: PMC6305848 DOI: 10.1262/jrd.2018-110] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Kisspeptin, identified as a natural ligand of GPR54 in 2001, is now considered as a master regulator of puberty and subsequent reproductive functions in mammals. Our previous studies using
Kiss1 knockout (KO) rats clearly demonstrated the indispensable role of kisspeptin in gonadotropin-releasing hormone (GnRH)/gonadotropin secretion. In addition, behavioral
analyses of Kiss1 KO rats revealed an organizational effect of kisspeptin on neural circuits controlling sexual behaviors. Our studies using transgenic mice carrying a
region-specific Kiss1 enhancer-driven reporter gene provided a clue as to the mechanism by which estrogen regulates Kiss1 expression in hypothalamic
kisspeptin neurons. Analyses of Kiss1 expression and gonadotropin secretion during the pubertal transition shed light on the mechanism triggering GnRH/gonadotropin secretion
at the onset of puberty in rats. Here, we summarize data obtained from the aforementioned studies and revisit the physiological roles of kisspeptin in the mechanism underlying reproductive
functions in mammals.
Collapse
Affiliation(s)
- Yoshihisa Uenoyama
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Naoko Inoue
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Kei-Ichiro Maeda
- Department of Veterinary Medical Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Hiroko Tsukamura
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| |
Collapse
|
27
|
Harter CJL, Kavanagh GS, Smith JT. The role of kisspeptin neurons in reproduction and metabolism. J Endocrinol 2018; 238:R173-R183. [PMID: 30042117 DOI: 10.1530/joe-18-0108] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Accepted: 06/13/2018] [Indexed: 02/06/2023]
Abstract
Kisspeptin is a neuropeptide with a critical role in the function of the hypothalamic-pituitary-gonadal (HPG) axis. Kisspeptin is produced by two major populations of neurons located in the hypothalamus, the rostral periventricular region of the third ventricle (RP3V) and arcuate nucleus (ARC). These neurons project to and activate gonadotrophin-releasing hormone (GnRH) neurons (acting via the kisspeptin receptor, Kiss1r) in the hypothalamus and stimulate the secretion of GnRH. Gonadal sex steroids stimulate kisspeptin neurons in the RP3V, but inhibit kisspeptin neurons in the ARC, which is the underlying mechanism for positive- and negative feedback respectively, and it is now commonly accepted that the ARC kisspeptin neurons act as the GnRH pulse generator. Due to kisspeptin's profound effect on the HPG axis, a focus of recent research has been on afferent inputs to kisspeptin neurons and one specific area of interest has been energy balance, which is thought to facilitate effects such as suppressing fertility in those with under- or severe over-nutrition. Alternatively, evidence is building for a direct role for kisspeptin in regulating energy balance and metabolism. Kiss1r-knockout (KO) mice exhibit increased adiposity and reduced energy expenditure. Although the mechanisms underlying these observations are currently unknown, Kiss1r is expressed in adipose tissue and potentially brown adipose tissue (BAT) and Kiss1rKO mice exhibit reduced energy expenditure. Recent studies are now looking at the effects of kisspeptin signalling on behaviour, with clinical evidence emerging of kisspeptin affecting sexual behaviour, further investigation of potential neuronal pathways are warranted.
Collapse
Affiliation(s)
- Campbell J L Harter
- School of Human SciencesThe University of Western Australia, Perth, Western Australia, Australia
| | - Georgia S Kavanagh
- School of Human SciencesThe University of Western Australia, Perth, Western Australia, Australia
| | - Jeremy T Smith
- School of Human SciencesThe University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
28
|
Moore AM, Coolen LM, Porter DT, Goodman RL, Lehman MN. KNDy Cells Revisited. Endocrinology 2018; 159:3219-3234. [PMID: 30010844 PMCID: PMC6098225 DOI: 10.1210/en.2018-00389] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 07/05/2018] [Indexed: 12/29/2022]
Abstract
In the past decade since kisspeptin/neurokinin B/dynorphin (KNDy) cells were first identified in the mammalian hypothalamus, a plethora of new research has emerged adding insights into the role of this neuronal population in reproductive neuroendocrine function, including the basis for GnRH pulse generation and the mechanisms underlying the steroid feedback control of GnRH secretion. In this mini-review, we provide an update of evidence regarding the roles of KNDy peptides and their postsynaptic receptors in producing episodic GnRH release and assess the relative contribution of KNDy neurons to the "GnRH pulse generator." In addition, we examine recent work investigating the role of KNDy neurons as mediators of steroid hormone negative feedback and review evidence for their involvement in the preovulatory GnRH/LH surge, taking into account species differences that exist among rodents, ruminants, and primates. Finally, we summarize emerging roles of KNDy neurons in other aspects of reproductive function and in nonreproductive functions and discuss critical unresolved questions in our understanding of KNDy neurobiology.
Collapse
Affiliation(s)
- Aleisha M Moore
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, Mississippi
| | - Lique M Coolen
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, Mississippi
- Department of Physics and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Danielle T Porter
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, Mississippi
| | - Robert L Goodman
- Department of Physiology, Pharmacology, and Neuroscience, West Virginia University, Morgantown, West Virginia
| | - Michael N Lehman
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
29
|
Okamura H, Yamamura T, Wakabayashi Y. Mapping of KNDy neurons and immunohistochemical analysis of the interaction between KNDy and substance P neural systems in goat. J Reprod Dev 2017; 63:571-580. [PMID: 29109352 PMCID: PMC5735268 DOI: 10.1262/jrd.2017-103] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
A population of neurons in the arcuate nucleus (ARC) coexpresses kisspeptin, neurokinin B (NKB), and dynorphin, and therefore they are referred to as KNDy neurons. It has been suggested that KNDy neurons participate in several brain functions, including the control of reproduction. The present study aimed to advance our understanding of the anatomy of the KNDy neural system. We first produced an antiserum against goat kisspeptin. After confirming its specificity, the antiserum was used to histochemically detect kisspeptin-positive signals. Using the colocalization of kisspeptin and NKB immunoreactivity as a marker for KNDy neurons, we mapped distributions of their cell somata and fibers in the whole brain (except the cerebellum) of ovariectomized (OVX) goats. KNDy neuronal somata were distributed throughout the ARC, and were particularly abundant in its caudal aspect. KNDy neuronal fibers projected into several areas within the septo-preoptic-hypothalamic continuum, such as the ARC, median eminence, medial preoptic nucleus, and bed nucleus of the stria terminalis. Kisspeptin immunoreactivity was not found outside of the continuum. We then addressed to the hypothesis that substance P (SP) is also involved in the KNDy neural system. Double-labeling immunohistochemistry for kisspeptin and SP revealed that KNDy neurons did not coexpress SP, but nearly all of the KNDy neuronal somata were surrounded by fibers containing SP in the OVX goats. The present results demonstrate anatomical evidence for a robust association between the KNDy and SP neural systems.
Collapse
Affiliation(s)
- Hiroaki Okamura
- Division of Animal Breeding and Reproduction Research, Institute of Livestock and Grassland Science, NARO, Ibaraki 305-0901, Japan
| | - Takashi Yamamura
- Division of Animal Breeding and Reproduction Research, Institute of Livestock and Grassland Science, NARO, Ibaraki 305-0901, Japan
| | - Yoshihiro Wakabayashi
- Division of Animal Breeding and Reproduction Research, Institute of Livestock and Grassland Science, NARO, Ibaraki 305-0901, Japan
| |
Collapse
|
30
|
Minabe S, Ieda N, Watanabe Y, Inoue N, Uenoyama Y, Maeda KI, Tsukamura H. Long-Term Neonatal Estrogen Exposure Causes Irreversible Inhibition of LH Pulses by Suppressing Arcuate Kisspeptin Expression via Estrogen Receptors α and β in Female Rodents. Endocrinology 2017; 158:2918-2929. [PMID: 28368450 DOI: 10.1210/en.2016-1144] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 03/22/2017] [Indexed: 11/19/2022]
Abstract
Exposure to estrogen during the developmental period causes reproductive dysfunction in mammals, because the developing brain is highly sensitive to estrogens. In the present study, we report that long-term exposure to supraphysiological doses of estrogen during the neonatal critical period causes irreversible suppression of Kiss1/kisspeptin expression in the arcuate nucleus (ARC) via estrogen receptor-alpha (ERα) and ERβ, resulting in reproductive dysfunction in female rats. Daily estradiol-benzoate (EB) administration from days 0 to 10 postpartum caused persistent vaginal diestrus in female rats. The female rats showed profound suppression of pulsatile luteinizing hormone (LH) release and ARC Kiss1/kisspeptin expression even after ovariectomy at adulthood. In contrast, female rats treated with a single EB injection at day 5 postpartum exhibited persistent vaginal estrus and showed comparable LH pulses and numbers of ARC Kiss1-expressing cells to vehicle-treated controls after ovariectomy at adulthood. Because the LH secretory response to exogenous kisspeptin was spared in female rats with neonatal long-term estrogen exposure, the LH pulse suppression was most probably due to ARC kisspeptin deficiency. Furthermore, neonatal estrogen might act through both ERα and ERβ, because EB exposure significantly reduced the number of ARC Kiss1-expressing cells in wild-type mice but not in ERα or ERβ knockout mice. Taken together, long-term exposure to supraphysiological doses of estrogen in the developing brain might cause defects in ARC kisspeptin neurons via ERα and ERβ, resulting in inhibition of pulsatile LH release and lack of estrous cyclicity.
Collapse
Affiliation(s)
- Shiori Minabe
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi 464-8601, Japan
| | - Nahoko Ieda
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi 464-8601, Japan
| | - Youki Watanabe
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi 464-8601, Japan
| | - Naoko Inoue
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi 464-8601, Japan
| | - Yoshihisa Uenoyama
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi 464-8601, Japan
| | - Kei-Ichiro Maeda
- Department of Veterinary Medical Sciences, University of Tokyo, Tokyo 113-8657, Japan
| | - Hiroko Tsukamura
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi 464-8601, Japan
| |
Collapse
|
31
|
Ikegami K, Minabe S, Ieda N, Goto T, Sugimoto A, Nakamura S, Inoue N, Oishi S, Maturana AD, Sanbo M, Hirabayashi M, Maeda KI, Tsukamura H, Uenoyama Y. Evidence of involvement of neurone-glia/neurone-neurone communications via gap junctions in synchronised activity of KNDy neurones. J Neuroendocrinol 2017; 29. [PMID: 28475285 DOI: 10.1111/jne.12480] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 04/21/2017] [Accepted: 05/01/2017] [Indexed: 11/27/2022]
Abstract
Pulsatile secretion of gonadotrophin-releasing hormone (GnRH)/luteinising hormone is indispensable for the onset of puberty and reproductive activities at adulthood in mammalian species. A cohort of neurones expressing three neuropeptides, namely kisspeptin, encoded by the Kiss1 gene, neurokinin B (NKB) and dynorphin A, localised in the hypothalamic arcuate nucleus (ARC), so-called KNDy neurones, comprises a putative intrinsic source of the GnRH pulse generator. Synchronous activity among KNDy neurones is considered to be required for pulsatile GnRH secretion. It has been reported that gap junctions play a key role in synchronising electrical activity in the central nervous system. Thus, we hypothesised that gap junctions are involved in the synchronised activities of KNDy neurones, which is induced by NKB-NK3R signalling. We determined the role of NKB-NK3R signalling in Ca2+ oscillation (an indicator of neuronal activities) of KNDy neurones and its synchronisation mechanism among KNDy neurones. Senktide, a selective agonist for NK3R, increased the frequency of Ca2+ oscillations in cultured Kiss1-GFP cells collected from the mediobasal hypothalamus of the foetal Kiss1-green fluorescent protein (GFP) mice. The senktide-induced Ca2+ oscillations were synchronised in the Kiss1-GFP and neighbouring glial cells. Confocal microscopy analysis of these cells, which have shown synchronised Ca2+ oscillations, revealed close contacts between Kiss1-GFP cells, as well as between Kiss1-GFP cells and glial cells. Dye coupling experiments suggest cell-to-cell communication through gap junctions between Kiss1-GFP cells and neighbouring glial cells. Connexin-26 and -37 mRNA were found in isolated ARC Kiss1 cells taken from adult female Kiss1-GFP transgenic mice. Furthermore, 18β-glycyrrhetinic acids and mefloquine, which are gap junction inhibitors, attenuated senktide-induced Ca2+ oscillations in Kiss1-GFP cells. Taken together, these results suggest that NKB-NK3R signalling enhances synchronised activities among neighbouring KNDy neurones, and that both neurone-neurone and neurone-glia communications via gap junctions possibly contribute to synchronised activities among KNDy neurones.
Collapse
Affiliation(s)
- K Ikegami
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - S Minabe
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - N Ieda
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - T Goto
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
- Centre for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Okazaki, Japan
| | - A Sugimoto
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - S Nakamura
- Department of Veterinary Medical Sciences, The University of Tokyo, Tokyo, Japan
| | - N Inoue
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - S Oishi
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - A D Maturana
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - M Sanbo
- Centre for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Okazaki, Japan
| | - M Hirabayashi
- Centre for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Okazaki, Japan
| | - K-I Maeda
- Department of Veterinary Medical Sciences, The University of Tokyo, Tokyo, Japan
| | - H Tsukamura
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Y Uenoyama
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| |
Collapse
|
32
|
d'Anglemont de Tassigny X, Jayasena C, Murphy KG, Dhillo WS, Colledge WH. Mechanistic insights into the more potent effect of KP-54 compared to KP-10 in vivo. PLoS One 2017; 12:e0176821. [PMID: 28464043 PMCID: PMC5413024 DOI: 10.1371/journal.pone.0176821] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 04/18/2017] [Indexed: 01/03/2023] Open
Abstract
Kisspeptins regulate the mammalian reproductive axis by stimulating release of gonadotrophin releasing hormone (GnRH). Different length kisspeptins (KP) are found of 54, 14, 13 or 10 amino-acids which share a common C-terminal 10-amino acid sequence. KP-54 and KP-10 have been widely used to stimulate the reproductive axis but data suggest that KP-54 and KP-10 are not equally effective at eliciting reproductive hormone secretion after peripheral delivery. To confirm this, we analysed the effect of systemic administration of KP-54 or KP-10 on luteinizing hormone (LH) secretion into the bloodstream of male mice. Plasma LH measurements 10 min or 2 hours after kisspeptin injection showed that KP-54 can sustain LH release far longer than KP-10, suggesting a differential mode of action of the two peptides. To investigate the mechanism for this, we evaluated the pharmacokinetics of the two peptides in vivo and their potential to cross the blood brain barrier (BBB). We found that KP-54 has a half-life of ~32 min in the bloodstream, while KP-10 has a half-life of ~4 min. To compensate for this difference in half-life, we repeated injections of KP-10 every 10 min over 1 hr but failed to reproduce the sustained rise in LH observed after a single KP-54 injection, suggesting that the failure of KP-10 to sustain LH release may not just be related to peptide clearance. We tested the ability of peripherally administered KP-54 and KP-10 to activate c-FOS in GnRH neurons behind the blood brain barrier (BBB) and found that only KP-54 could do this. These data are consistent with KP-54 being able to cross the BBB and suggest that KP10 may be less able to do so.
Collapse
Affiliation(s)
- Xavier d'Anglemont de Tassigny
- Reproductive Physiology Group, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Channa Jayasena
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London at Hammersmith Campus, Commonwealth Building, London, United Kingdom
| | - Kevin G. Murphy
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London at Hammersmith Campus, Commonwealth Building, London, United Kingdom
| | - Waljit S. Dhillo
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London at Hammersmith Campus, Commonwealth Building, London, United Kingdom
| | - William H. Colledge
- Reproductive Physiology Group, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
- * E-mail:
| |
Collapse
|
33
|
Spike and Neuropeptide-Dependent Mechanisms Control GnRH Neuron Nerve Terminal Ca 2+ over Diverse Time Scales. J Neurosci 2017; 37:3342-3351. [PMID: 28235895 DOI: 10.1523/jneurosci.2925-16.2017] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Revised: 02/14/2017] [Accepted: 02/17/2017] [Indexed: 01/30/2023] Open
Abstract
Fast cell-to-cell communication in the brain is achieved by action potential-dependent synaptic release of neurotransmitters. The fast kinetics of transmitter release are determined by transient Ca2+ elevations in presynaptic nerve terminals. Neuromodulators have previously been shown to regulate transmitter release by inhibiting presynaptic Ca2+ influx. Few studies to date have demonstrated the opposite, that is, neuromodulators directly driving presynaptic Ca2+ rises and increases in nerve terminal excitability. Here we use GCaMP Ca2+ imaging in brain slices from mice to address how nerve terminal Ca2+ is controlled in gonadotropin-releasing hormone (GnRH) neurons via action potentials and neuromodulators. Single spikes and bursts of action potentials evoked fast, voltage-gated Ca2+ channel-dependent Ca2+ elevations. In contrast, brief exposure to the neuropeptide kisspeptin-evoked long-lasting Ca2+ plateaus that persisted for tens of minutes. Neuropeptide-mediated Ca2+ elevations were independent of action potentials, requiring Ca2+ entry via voltage-gated Ca2+ channels and transient receptor potential channels in addition to release from intracellular store mechanisms. Together, these data reveal that neuromodulators can exert powerful and long-lasting regulation of nerve terminal Ca2+ independently from actions at the soma. Thus, GnRH nerve terminal function is controlled over disparate timescales via both classical spike-dependent and nonclassical neuropeptide-dependent mechanisms.SIGNIFICANCE STATEMENT Nerve terminals are highly specialized regions of a neuron where neurotransmitters and neurohormones are released. Many neuroendocrine neurons release neurohormones in long-duration bursts of secretion. To understand how this is achieved, we have performed live Ca2+ imaging in the nerve terminals of gonadotropin-releasing hormone neurons. We find that bursts of action potentials and local neuropeptide signals are both capable of evoking large increases in nerve terminal Ca2+ Increases in Ca2+ driven by spike bursts last seconds; however, the increases in nerve terminal Ca2+ driven by neuropeptides can persist for tens of minutes. These findings reveal new mechanisms by which neuroendocrine nerve terminal Ca2+ can be controlled in the brain.
Collapse
|
34
|
Sukhbaatar U, Kanasaki H, Mijiddorj T, Oride A, Hara T, Yamada T, Kyo S. Expression of GnRH and Kisspeptin in Primary Cultures of Fetal Rat Brain. Reprod Sci 2016; 24:227-233. [PMID: 27313117 DOI: 10.1177/1933719116653679] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Genetic studies in humans or in vivo studies using animals have shown that kisspeptin released from the hypothalamus controls secretion of gonadotropin-releasing hormone (GnRH) from GnRH neurons, and subsequently GnRH induces gonadotropin secretion from the anterior pituitary. Kisspeptin did not stimulate GnRH expression in the GnRH-producing cell line GT1-7. Thus, we cultured GnRH and kisspeptin neurons from whole fetal rat brain and examined the regulation of GnRH and kisspeptin. Expression of GnRH messenger RNA (mRNA) was unchanged by estradiol (E2) treatment in these primary cultures. In contrast, kisspeptin mRNA expression was increased 2.00 ± 0.23-fold by E2 treatment. When these cultures were stimulated by kisspeptin-10, GnRH mRNA was significantly increased up to 1.51 ± 0.35-fold. Expression of GnRH mRNA was also stimulated 1.84 ± 0.33-fold by GnRH itself. Interestingly, kisspeptin mRNA was significantly increased up to 2.43 ± 0.40-fold by kisspeptin alone. In addition, kisspeptin mRNA expression was significantly increased by stimulation with GnRH (1.46 ± 0.21-fold). Our observations demonstrated that kisspeptin, but not GnRH, was upregulated by E2 and that kisspeptin stimulates GnRH mRNA expression in primary cultures of whole fetal rat brain. Furthermore, GnRH and kisspeptin stimulate their own neurons to produce GnRH or kisspeptin, respectively.
Collapse
Affiliation(s)
- Unurjargal Sukhbaatar
- 1 Department of Obstetrics and Gynecology, Faculty of Medicine, Shimane University School of Medicine, Izumo, Shimane, Japan
| | - Haruhiko Kanasaki
- 1 Department of Obstetrics and Gynecology, Faculty of Medicine, Shimane University School of Medicine, Izumo, Shimane, Japan
| | - Tselmeg Mijiddorj
- 1 Department of Obstetrics and Gynecology, Faculty of Medicine, Shimane University School of Medicine, Izumo, Shimane, Japan
| | - Aki Oride
- 1 Department of Obstetrics and Gynecology, Faculty of Medicine, Shimane University School of Medicine, Izumo, Shimane, Japan
| | - Tomomi Hara
- 1 Department of Obstetrics and Gynecology, Faculty of Medicine, Shimane University School of Medicine, Izumo, Shimane, Japan
| | - Takaya Yamada
- 2 Department of Experimental Animals, Interdisciplinary Center for Science Research, Organization for Research, Shimane University, Izumo, Shimane, Japan
| | - Satoru Kyo
- 1 Department of Obstetrics and Gynecology, Faculty of Medicine, Shimane University School of Medicine, Izumo, Shimane, Japan
| |
Collapse
|
35
|
Uenoyama Y, Pheng V, Tsukamura H, Maeda KI. The roles of kisspeptin revisited: inside and outside the hypothalamus. J Reprod Dev 2016; 62:537-545. [PMID: 27478063 PMCID: PMC5177970 DOI: 10.1262/jrd.2016-083] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Kisspeptin, encoded by KISS1/Kiss1 gene, is now considered a master regulator of reproductive functions in mammals owing to
its involvement in the direct activation of gonadotropin-releasing hormone (GnRH) neurons after binding to its cognate receptor, GPR54. Ever since the discovery
of kisspeptin, intensive studies on hypothalamic expression of KISS1/Kiss1 and on physiological roles of hypothalamic
kisspeptin neurons have provided clues as to how the brain controls sexual maturation at the onset of puberty and subsequent reproductive performance in
mammals. Additionally, emerging evidence indicates the potential involvement of extra-hypothalamic kisspeptin in reproductive functions. Here, we summarize data
regarding kisspeptin inside and outside the hypothalamus and revisit the physiological roles of central and peripheral kisspeptins in the reproductive functions
of mammals.
Collapse
Affiliation(s)
- Yoshihisa Uenoyama
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | | | | | | |
Collapse
|
36
|
Hassaneen A, Naniwa Y, Suetomi Y, Matsuyama S, Kimura K, Ieda N, Inoue N, Uenoyama Y, Tsukamura H, Maeda KI, Matsuda F, Ohkura S. Immunohistochemical characterization of the arcuate kisspeptin/neurokinin B/dynorphin (KNDy) and preoptic kisspeptin neuronal populations in the hypothalamus during the estrous cycle in heifers. J Reprod Dev 2016; 62:471-477. [PMID: 27349533 PMCID: PMC5081734 DOI: 10.1262/jrd.2016-075] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Elucidating the physiological mechanisms that control reproduction is an obvious strategy for improving the fertility of cattle and developing new agents to
control reproductive functions. The present study aimed to identify kisspeptin neurons in the bovine hypothalamus, clarifying that a central mechanism is also
present in the cattle brain, as kisspeptin is known to play an important role in the stimulation of gonadotropin-releasing hormone (GnRH)/gonadotropin secretion
in other mammals. To characterize kisspeptin neurons in the bovine hypothalamus, the co-localizations of kisspeptin and neurokinin B (NKB) or kisspeptin and
dynorphin A (Dyn) were examined. Hypothalamic tissue was collected from Japanese Black or Japanese Black × Holstein crossbred cows during the follicular and
luteal phases. Brain sections, including the arcuate nucleus (ARC) and the preoptic area (POA), were dual immunostained with kisspeptin and either NKB or Dyn.
In the ARC, both NKB and Dyn were co-localized in kisspeptin neurons during both the follicular and luteal phases, demonstrating the presence of
kisspeptin/NKB/Dyn-containing neurons, referred to as KNDy neurons, in cows. In the POA, no co-localization of kisspeptin with either NKB or Dyn was detected.
Kisspeptin expression in the follicular phase was higher than that in the luteal phase, suggesting that kisspeptin expression in the POA is positively
controlled by estrogen in cows. The kisspeptin neuronal populations in the ARC and POA likely play important roles in regulating the GnRH pulse and surge,
respectively, in cows.
Collapse
Affiliation(s)
- A Hassaneen
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Liu X, Herbison AE. Kisspeptin Regulation of Neuronal Activity throughout the Central Nervous System. Endocrinol Metab (Seoul) 2016; 31:193-205. [PMID: 27246282 PMCID: PMC4923402 DOI: 10.3803/enm.2016.31.2.193] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 04/14/2016] [Accepted: 04/22/2016] [Indexed: 12/30/2022] Open
Abstract
Kisspeptin signaling at the gonadotropin-releasing hormone (GnRH) neuron is now relatively well characterized and established as being critical for the neural control of fertility. However, kisspeptin fibers and the kisspeptin receptor (KISS1R) are detected throughout the brain suggesting that kisspeptin is involved in regulating the activity of multiple neuronal circuits. We provide here a review of kisspeptin actions on neuronal populations throughout the brain including the magnocellular oxytocin and vasopressin neurons, and cells within the arcuate nucleus, hippocampus, and amygdala. The actions of kisspeptin in these brain regions are compared to its effects upon GnRH neurons. Two major themes arise from this analysis. First, it is apparent that kisspeptin signaling through KISS1R at the GnRH neuron is a unique, extremely potent form or neurotransmission whereas kisspeptin actions through KISS1R in other brain regions exhibit neuromodulatory actions typical of other neuropeptides. Second, it is becoming increasingly likely that kisspeptin acts as a neuromodulator not only through KISS1R but also through other RFamide receptors such as the neuropeptide FF receptors (NPFFRs). We suggest likely locations of kisspeptin signaling through NPFFRs but note that only limited tools are presently available for examining kisspeptin cross-signaling within the RFamide family of neuropeptides.
Collapse
Affiliation(s)
- Xinhuai Liu
- Centre for Neuroendocrinology, Department of Physiology, University of Otago School of Medical Sciences, Dunedin, New Zealand
| | - Allan E Herbison
- Centre for Neuroendocrinology, Department of Physiology, University of Otago School of Medical Sciences, Dunedin, New Zealand.
| |
Collapse
|
38
|
A synthetic kisspeptin analog that triggers ovulation and advances puberty. Sci Rep 2016; 6:26908. [PMID: 27245315 PMCID: PMC4887910 DOI: 10.1038/srep26908] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 05/09/2016] [Indexed: 12/26/2022] Open
Abstract
The neuropeptide kisspeptin and its receptor, KiSS1R, govern the reproductive timeline of mammals by triggering puberty onset and promoting ovulation by stimulating gonadotrophin-releasing hormone (GnRH) secretion. To overcome the drawback of kisspeptin short half-life we designed kisspeptin analogs combining original modifications, triazole peptidomimetic and albumin binding motif, to reduce proteolytic degradation and to slow down renal clearance, respectively. These analogs showed improved in vitro potency and dramatically enhanced pharmacodynamics. When injected intramuscularly into ewes (15 nmol/ewe) primed with a progestogen, the best analog (compound 6, C6) induced synchronized ovulations in both breeding and non-breeding seasons. Ovulations were fertile as demonstrated by the delivery of lambs at term. C6 was also fully active in both female and male mice but was completely inactive in KiSS1R KO mice. Electrophysiological recordings of GnRH neurons from brain slices of GnRH-GFP mice indicated that C6 exerted a direct excitatory action on GnRH neurons. Finally, in prepubertal female mice daily injections (0.3 nmol/mouse) for five days significantly advanced puberty. C6 ability to trigger ovulation and advance puberty demonstrates that kisspeptin analogs may find application in the management of livestock reproduction and opens new possibilities for the treatment of reproductive disorders in humans.
Collapse
|
39
|
Tanco VM, Whitlock BK, Jones MA, Wilborn RR, Brandebourg TD, Foradori CD. Distribution and regulation of gonadotropin-releasing hormone, kisspeptin, RF-amide related peptide-3, and dynorphin in the bovine hypothalamus. PeerJ 2016; 4:e1833. [PMID: 27014517 PMCID: PMC4806599 DOI: 10.7717/peerj.1833] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 03/01/2016] [Indexed: 01/06/2023] Open
Abstract
Recent work has led to the hypothesis that kisspeptin/neurokinin B/dynorphin (KNDy) neurons in the arcuate nucleus (ARC) play a key role in gonadotropin-releasing hormone (GnRH) pulse generation and gonadal steroid feedback, with kisspeptin driving GnRH release and neurokinin B and dynorphin acting as pulse start and stop signals, respectively. A separate cell group, expressing RFamide-related peptide-3 (RFRP-3) has been shown to be a primary inhibitor of GnRH release. Very little is known regarding these cell groups in the bovine. In this study, we examined the relative immunoreactivity of kisspeptin, dynorphin, and RFRP-3 and their possible connectivity to GnRH neurons in the hypothalami of periestrus and diestrus bovine. While GnRH and RFRP-3 immunoreactivity were unchanged, kisspeptin and dynorphin immunoreactivity levels varied in relation to plasma progesterone concentrations and estrous status. Animals with higher plasma progesterone concentrations in diestrus had lower kisspeptin and increased dynorphin immunoreactivity in the ARC. The percentage of GnRH cells with kisspeptin or RFRP-3 fibers in close apposition did not differ between estrous stages. However, the proportions of GnRH cells with kisspeptin or RFRP-3 contacts (∼49.8% and ∼31.3%, respectively) suggest direct communication between kisspeptin and RFRP-3 cells to GnRH cells in the bovine. The data produced in this work support roles for kisspeptin and dynorphin, within the KNDy neural network, in controlling GnRH release over the ovarian cycle and conveying progesterone-negative feedback onto GnRH neurons in the bovine.
Collapse
Affiliation(s)
- Valeria M Tanco
- Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee-Knoxville , Knoxville, TN , United States
| | - Brian K Whitlock
- Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee-Knoxville , Knoxville, TN , United States
| | - Melaney A Jones
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University , Auburn, AL , United States
| | - Robyn R Wilborn
- Department of Clinical Sciences, College of Veterinary Medicine, Auburn University , Auburn, AL , United States
| | - Terry D Brandebourg
- Department of Animal Sciences, College of Agriculture, Auburn University , Auburn, AL , United States
| | - Chad D Foradori
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University , Auburn, AL , United States
| |
Collapse
|
40
|
Fernandois D, Na E, Cuevas F, Cruz G, Lara HE, Paredes AH. Kisspeptin is involved in ovarian follicular development during aging in rats. J Endocrinol 2016; 228:161-70. [PMID: 26698566 DOI: 10.1530/joe-15-0429] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/22/2015] [Indexed: 11/08/2022]
Abstract
We have previously reported that kisspeptin (KP) may be under the control of the sympathetic innervation of the ovary. Considering that the sympathetic activity of the ovary increases with aging, it is possible that ovarian KP also increases during this period and participates in follicular development. To evaluate this possibility, we determined ovarian KP expression and its action on follicular development during reproductive aging in rats. We measured ovarian KP mRNA and protein levels in 6-, 8-, 10- and 12-month-old rats. To evaluate follicular developmental changes, intraovarian administration of KP or its antagonist, peptide 234 (P234), was performed using a mini-osmotic pump, and to evaluate FSH receptor (FSHR) changes in the senescent ovary, we stimulated cultured ovaries with KP, P234 and isoproterenol (ISO). Our results shows that KP expression in the ovary was increased in 10- and 12-month-old rats compared with 6-month-old rats, and this increase in KP was strongly correlated with the increase in ovarian norepinephrine observed with aging. The administration of KP produced an increase in corpora lutea and type III follicles in 6- and 10-month-old rats, which was reversed by P234 administration at 10 months. In addition, KP decreased the number and size of antral follicles in 6- and 10-month-old rats, while P234 administration produced an increase in these structures at the same ages. In ovarian cultures KP prevented the induction of FSHR by ISO. These results suggest that intraovarian KP negatively participates in the acquisition of FSHR, indicating a local role in the regulation of follicular development and ovulation during reproductive aging.
Collapse
Affiliation(s)
- D Fernandois
- Laboratory of NeurobiochemistryDepartment of Biochemistry and Molecular Biology, Faculty of Chemistry and Pharmaceutical Sciences, Universidad de Chile, Santiago 8380492, ChileLaboratorio de alteraciones Reproductivas y MetabólicasFacultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - E Na
- Laboratory of NeurobiochemistryDepartment of Biochemistry and Molecular Biology, Faculty of Chemistry and Pharmaceutical Sciences, Universidad de Chile, Santiago 8380492, ChileLaboratorio de alteraciones Reproductivas y MetabólicasFacultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - F Cuevas
- Laboratory of NeurobiochemistryDepartment of Biochemistry and Molecular Biology, Faculty of Chemistry and Pharmaceutical Sciences, Universidad de Chile, Santiago 8380492, ChileLaboratorio de alteraciones Reproductivas y MetabólicasFacultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - G Cruz
- Laboratory of NeurobiochemistryDepartment of Biochemistry and Molecular Biology, Faculty of Chemistry and Pharmaceutical Sciences, Universidad de Chile, Santiago 8380492, ChileLaboratorio de alteraciones Reproductivas y MetabólicasFacultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - H E Lara
- Laboratory of NeurobiochemistryDepartment of Biochemistry and Molecular Biology, Faculty of Chemistry and Pharmaceutical Sciences, Universidad de Chile, Santiago 8380492, ChileLaboratorio de alteraciones Reproductivas y MetabólicasFacultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - A H Paredes
- Laboratory of NeurobiochemistryDepartment of Biochemistry and Molecular Biology, Faculty of Chemistry and Pharmaceutical Sciences, Universidad de Chile, Santiago 8380492, ChileLaboratorio de alteraciones Reproductivas y MetabólicasFacultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| |
Collapse
|
41
|
Iijima N, Takumi K, Matsumoto K, Ozawa H. Visualization of Kisspeptin Binding to Rat Hypothalamic Neurons. Acta Histochem Cytochem 2015; 48:179-84. [PMID: 26855450 PMCID: PMC4731855 DOI: 10.1267/ahc.15017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 10/09/2015] [Indexed: 11/30/2022] Open
Abstract
The neuropeptide kisspeptin plays an important role in fertility and the onset of puberty, stimulating gonadotropin-releasing hormone (GnRH) neurons to activate the hypothalamic–pituitary–gonadal axis. Several studies have demonstrated a morphological interaction between kisspeptin- and GnRH-expressing neurons; however, few have addressed the interaction of kisspeptin with other neuronal subtypes. We recently showed that fibers immunoreactive for kisspeptin were densely distributed in the dorsal part of the arcuate nucleus. These fibers were found in close proximity to GnRH and tuberoinfundibular dopamine (TIDA) neurons. In the present study, using biotinylated kisspeptin, we established a visualization method for identifying kisspeptin binding sites on TIDA neurons. Biotinylated kisspeptin bound to the cell bodies of TIDA neurons and surrounding fibers, suggesting that TIDA neurons express sites of action for kisspeptin. Our assay also detected biotinylation signals from kisspeptin binding to GnRH fibers in the median eminence, but not to cell bodies of GnRH neurons in the medial preoptic area. Positive signals were completely eliminated by addition of excess non-labeled kisspeptin. This method enabled us to detect kisspeptin binding sites on specific neural structures and neuronal fibers.
Collapse
Affiliation(s)
- Norio Iijima
- Department of Anatomy and Neurobiology, Graduate School of Medicine, Nippon Medical School
| | - Ken Takumi
- Department of Anatomy and Neurobiology, Graduate School of Medicine, Nippon Medical School
| | - Keisuke Matsumoto
- Department of Anatomy and Neurobiology, Graduate School of Medicine, Nippon Medical School
| | - Hitoshi Ozawa
- Department of Anatomy and Neurobiology, Graduate School of Medicine, Nippon Medical School
| |
Collapse
|
42
|
Murakawa H, Iwata K, Takeshita T, Ozawa H. Immunoelectron microscopic observation of the subcellular localization of kisspeptin, neurokinin B and dynorphin A in KNDy neurons in the arcuate nucleus of the female rat. Neurosci Lett 2015; 612:161-166. [PMID: 26679227 DOI: 10.1016/j.neulet.2015.12.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 11/25/2015] [Accepted: 12/04/2015] [Indexed: 01/31/2023]
Abstract
KNDy neurons are named for their co-expression of three neuropeptides, kisspeptin, neurokinin B (NKB) and dynorphin A (DynA). These cells, located in the hypothalamic arcuate nucleus (ARC), are associated with generation of gonadotropin-releasing hormone (GnRH)/luteinizing hormone (LH) pulses to control follicular growth and steroidogenesis. However, subcellular sorting mechanisms for secretory vesicles containing these neuropeptides have not been elucidated. In this study, we analyzed the localization pattern of kisspeptin, NKB and DynA in the ARC of the ovariectomized rat immediately treated with estrogen using immunoelectron microscopy. First, we identified neuropeptides by dual-labeled fluorescence immunohistochemistry, with results indicating all three neuropeptides co-express within individual ARC cells in female rats. Next, we investigated the subcellular localization pattern of kisspeptin, NKB, and/or DynA using post-embedding double immunoelectron microscopy, indicating that each type of neuropeptide is contained within separate and individual neurosecretory vesicles. This suggests sorting and packaging of kisspeptin, NKB and DynA is differentially regulated within KNDy neurons. Our findings facilitate understanding of regulatory mechanisms underlying kisspeptin secretion in KNDy neurons, and generation of GnRH/LH pulses induced by kisspeptin in the ARC.
Collapse
Affiliation(s)
- Hiroko Murakawa
- Department of Anatomy and Neurology, Graduate School of Medicine, Nippon Medical School, Bunkyo-ku, Tokyo 113-8602, Japan; Department of Reproductive Medicine, Perinatology and Gynecologic Oncology, Graduate School of Medicine, Nippon Medical School, Bunkyo-ku, Tokyo 113-8602, Japan
| | - Kinuyo Iwata
- Department of Anatomy and Neurology, Graduate School of Medicine, Nippon Medical School, Bunkyo-ku, Tokyo 113-8602, Japan
| | - Toshiyuki Takeshita
- Department of Reproductive Medicine, Perinatology and Gynecologic Oncology, Graduate School of Medicine, Nippon Medical School, Bunkyo-ku, Tokyo 113-8602, Japan
| | - Hitoshi Ozawa
- Department of Anatomy and Neurology, Graduate School of Medicine, Nippon Medical School, Bunkyo-ku, Tokyo 113-8602, Japan.
| |
Collapse
|
43
|
Helena CV, Toporikova N, Kalil B, Stathopoulos AM, Pogrebna VV, Carolino RO, Anselmo-Franci JA, Bertram R. KNDy Neurons Modulate the Magnitude of the Steroid-Induced Luteinizing Hormone Surges in Ovariectomized Rats. Endocrinology 2015; 156:4200-13. [PMID: 26302111 PMCID: PMC4606747 DOI: 10.1210/en.2015-1070] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Kisspeptin is the most potent stimulator of LH release. There are two kisspeptin neuronal populations in the rodent brain: in the anteroventral periventricular nucleus (AVPV) and in the arcuate nucleus. The arcuate neurons coexpress kisspeptin, neurokinin B, and dynorphin and are called KNDy neurons. Because estradiol increases kisspeptin expression in the AVPV whereas it inhibits KNDy neurons, AVPV and KNDy neurons have been postulated to mediate the positive and negative feedback effects of estradiol on LH secretion, respectively. Yet the role of KNDy neurons during the positive feedback is not clear. In this study, ovariectomized rats were microinjected bilaterally into the arcuate nucleus with a saporin-conjugated neurokinin B receptor agonist for targeted ablation of approximately 70% of KNDy neurons. In oil-treated animals, ablation of KNDy neurons impaired the rise in LH after ovariectomy and kisspeptin content in both populations. In estradiol-treated animals, KNDy ablation did not influence the negative feedback of steroids during the morning. Surprisingly, KNDy ablation increased the steroid-induced LH surges, accompanied by an increase of kisspeptin content in the AVPV. This increase seems to be due to lack of dynorphin input from KNDy neurons to the AVPV as the following: 1) microinjections of a dynorphin antagonist into the AVPV significantly increased the LH surge in estradiol-treated rats, similar to KNDy ablation, and 2) intra-AVPV microinjections of dynorphin in KNDy-ablated rats restored LH surge levels. Our results suggest that KNDy neurons provide inhibition to AVPV kisspeptin neurons through dynorphin and thus regulate the amplitude of the steroid-induced LH surges.
Collapse
Affiliation(s)
- Cleyde V Helena
- Program in Neuroscience and Department of Mathematics (C.V.H., R.B.) and Program in Neuroscience and Department of Biology (A.M.S.), Florida State University, Tallahassee, Florida 32306; Department of Biology (N.T., V.V.P.), Washington and Lee University, Lexington, Virginia 24450; and Department of Physiology (B.K.), Medical School, and Department of Morphology, Stomatology, and Physiology (R.O.C., J.A.A.-F.), School of Dentistry, University of São Paulo, Ribeirão Preto 14040-900, SP, Brazil
| | - Natalia Toporikova
- Program in Neuroscience and Department of Mathematics (C.V.H., R.B.) and Program in Neuroscience and Department of Biology (A.M.S.), Florida State University, Tallahassee, Florida 32306; Department of Biology (N.T., V.V.P.), Washington and Lee University, Lexington, Virginia 24450; and Department of Physiology (B.K.), Medical School, and Department of Morphology, Stomatology, and Physiology (R.O.C., J.A.A.-F.), School of Dentistry, University of São Paulo, Ribeirão Preto 14040-900, SP, Brazil
| | - Bruna Kalil
- Program in Neuroscience and Department of Mathematics (C.V.H., R.B.) and Program in Neuroscience and Department of Biology (A.M.S.), Florida State University, Tallahassee, Florida 32306; Department of Biology (N.T., V.V.P.), Washington and Lee University, Lexington, Virginia 24450; and Department of Physiology (B.K.), Medical School, and Department of Morphology, Stomatology, and Physiology (R.O.C., J.A.A.-F.), School of Dentistry, University of São Paulo, Ribeirão Preto 14040-900, SP, Brazil
| | - Andrea M Stathopoulos
- Program in Neuroscience and Department of Mathematics (C.V.H., R.B.) and Program in Neuroscience and Department of Biology (A.M.S.), Florida State University, Tallahassee, Florida 32306; Department of Biology (N.T., V.V.P.), Washington and Lee University, Lexington, Virginia 24450; and Department of Physiology (B.K.), Medical School, and Department of Morphology, Stomatology, and Physiology (R.O.C., J.A.A.-F.), School of Dentistry, University of São Paulo, Ribeirão Preto 14040-900, SP, Brazil
| | - Veronika V Pogrebna
- Program in Neuroscience and Department of Mathematics (C.V.H., R.B.) and Program in Neuroscience and Department of Biology (A.M.S.), Florida State University, Tallahassee, Florida 32306; Department of Biology (N.T., V.V.P.), Washington and Lee University, Lexington, Virginia 24450; and Department of Physiology (B.K.), Medical School, and Department of Morphology, Stomatology, and Physiology (R.O.C., J.A.A.-F.), School of Dentistry, University of São Paulo, Ribeirão Preto 14040-900, SP, Brazil
| | - Ruither O Carolino
- Program in Neuroscience and Department of Mathematics (C.V.H., R.B.) and Program in Neuroscience and Department of Biology (A.M.S.), Florida State University, Tallahassee, Florida 32306; Department of Biology (N.T., V.V.P.), Washington and Lee University, Lexington, Virginia 24450; and Department of Physiology (B.K.), Medical School, and Department of Morphology, Stomatology, and Physiology (R.O.C., J.A.A.-F.), School of Dentistry, University of São Paulo, Ribeirão Preto 14040-900, SP, Brazil
| | - Janete A Anselmo-Franci
- Program in Neuroscience and Department of Mathematics (C.V.H., R.B.) and Program in Neuroscience and Department of Biology (A.M.S.), Florida State University, Tallahassee, Florida 32306; Department of Biology (N.T., V.V.P.), Washington and Lee University, Lexington, Virginia 24450; and Department of Physiology (B.K.), Medical School, and Department of Morphology, Stomatology, and Physiology (R.O.C., J.A.A.-F.), School of Dentistry, University of São Paulo, Ribeirão Preto 14040-900, SP, Brazil
| | - Richard Bertram
- Program in Neuroscience and Department of Mathematics (C.V.H., R.B.) and Program in Neuroscience and Department of Biology (A.M.S.), Florida State University, Tallahassee, Florida 32306; Department of Biology (N.T., V.V.P.), Washington and Lee University, Lexington, Virginia 24450; and Department of Physiology (B.K.), Medical School, and Department of Morphology, Stomatology, and Physiology (R.O.C., J.A.A.-F.), School of Dentistry, University of São Paulo, Ribeirão Preto 14040-900, SP, Brazil
| |
Collapse
|
44
|
Yin W, Maguire SM, Pham B, Garcia AN, Dang NV, Liang J, Wolfe A, Hofmann HA, Gore AC. Testing the Critical Window Hypothesis of Timing and Duration of Estradiol Treatment on Hypothalamic Gene Networks in Reproductively Mature and Aging Female Rats. Endocrinology 2015; 156:2918-33. [PMID: 26018250 PMCID: PMC4511137 DOI: 10.1210/en.2015-1032] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 05/05/2015] [Indexed: 12/12/2022]
Abstract
At menopause, the dramatic loss of ovarian estradiol (E2) necessitates the adaptation of estrogen-sensitive neurons in the hypothalamus to an estrogen-depleted environment. We developed a rat model to test the "critical window" hypothesis of the effects of timing and duration of E2 treatment after deprivation on the hypothalamic neuronal gene network in the arcuate nucleus and the medial preoptic area. Rats at 2 ages (reproductively mature or aging) were ovariectomized and given E2 or vehicle replacement regimes of differing timing and duration. Using a 48-gene quantitative low-density PCR array and weighted gene coexpression network analysis, we identified gene modules differentially regulated by age, timing, and duration of E2 treatment. Of particular interest, E2 status differentially affected suites of genes in the hypothalamus involved in energy balance, circadian rhythms, and reproduction. In fact, E2 status was the dominant factor in determining gene modules and hormone levels; age, timing, and duration had more subtle effects. Our results highlight the plasticity of hypothalamic neuroendocrine systems during reproductive aging and its surprising ability to adapt to diverse E2 replacement regimes.
Collapse
Affiliation(s)
- Weiling Yin
- Division of Pharmacology and Toxicology (W.Y., B.P., N.-V.D., J.L., A.C.G.), Departments of Integrative Biology (S.M.M., H.A.H.) and Psychology (A.N.G., A.C.G.), and Institute for Neuroscience (H.A.H., A.C.G.), The University of Texas at Austin, Austin, Texas 78712; and Johns Hopkins University School of Medicine (A.W.), Baltimore, Maryland 21287
| | - Sean M Maguire
- Division of Pharmacology and Toxicology (W.Y., B.P., N.-V.D., J.L., A.C.G.), Departments of Integrative Biology (S.M.M., H.A.H.) and Psychology (A.N.G., A.C.G.), and Institute for Neuroscience (H.A.H., A.C.G.), The University of Texas at Austin, Austin, Texas 78712; and Johns Hopkins University School of Medicine (A.W.), Baltimore, Maryland 21287
| | - Brian Pham
- Division of Pharmacology and Toxicology (W.Y., B.P., N.-V.D., J.L., A.C.G.), Departments of Integrative Biology (S.M.M., H.A.H.) and Psychology (A.N.G., A.C.G.), and Institute for Neuroscience (H.A.H., A.C.G.), The University of Texas at Austin, Austin, Texas 78712; and Johns Hopkins University School of Medicine (A.W.), Baltimore, Maryland 21287
| | - Alexandra N Garcia
- Division of Pharmacology and Toxicology (W.Y., B.P., N.-V.D., J.L., A.C.G.), Departments of Integrative Biology (S.M.M., H.A.H.) and Psychology (A.N.G., A.C.G.), and Institute for Neuroscience (H.A.H., A.C.G.), The University of Texas at Austin, Austin, Texas 78712; and Johns Hopkins University School of Medicine (A.W.), Baltimore, Maryland 21287
| | - Nguyen-Vy Dang
- Division of Pharmacology and Toxicology (W.Y., B.P., N.-V.D., J.L., A.C.G.), Departments of Integrative Biology (S.M.M., H.A.H.) and Psychology (A.N.G., A.C.G.), and Institute for Neuroscience (H.A.H., A.C.G.), The University of Texas at Austin, Austin, Texas 78712; and Johns Hopkins University School of Medicine (A.W.), Baltimore, Maryland 21287
| | - Jingya Liang
- Division of Pharmacology and Toxicology (W.Y., B.P., N.-V.D., J.L., A.C.G.), Departments of Integrative Biology (S.M.M., H.A.H.) and Psychology (A.N.G., A.C.G.), and Institute for Neuroscience (H.A.H., A.C.G.), The University of Texas at Austin, Austin, Texas 78712; and Johns Hopkins University School of Medicine (A.W.), Baltimore, Maryland 21287
| | - Andrew Wolfe
- Division of Pharmacology and Toxicology (W.Y., B.P., N.-V.D., J.L., A.C.G.), Departments of Integrative Biology (S.M.M., H.A.H.) and Psychology (A.N.G., A.C.G.), and Institute for Neuroscience (H.A.H., A.C.G.), The University of Texas at Austin, Austin, Texas 78712; and Johns Hopkins University School of Medicine (A.W.), Baltimore, Maryland 21287
| | - Hans A Hofmann
- Division of Pharmacology and Toxicology (W.Y., B.P., N.-V.D., J.L., A.C.G.), Departments of Integrative Biology (S.M.M., H.A.H.) and Psychology (A.N.G., A.C.G.), and Institute for Neuroscience (H.A.H., A.C.G.), The University of Texas at Austin, Austin, Texas 78712; and Johns Hopkins University School of Medicine (A.W.), Baltimore, Maryland 21287
| | - Andrea C Gore
- Division of Pharmacology and Toxicology (W.Y., B.P., N.-V.D., J.L., A.C.G.), Departments of Integrative Biology (S.M.M., H.A.H.) and Psychology (A.N.G., A.C.G.), and Institute for Neuroscience (H.A.H., A.C.G.), The University of Texas at Austin, Austin, Texas 78712; and Johns Hopkins University School of Medicine (A.W.), Baltimore, Maryland 21287
| |
Collapse
|
45
|
Yip SH, Boehm U, Herbison AE, Campbell RE. Conditional Viral Tract Tracing Delineates the Projections of the Distinct Kisspeptin Neuron Populations to Gonadotropin-Releasing Hormone (GnRH) Neurons in the Mouse. Endocrinology 2015; 156:2582-94. [PMID: 25856430 DOI: 10.1210/en.2015-1131] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Kisspeptin neurons play an essential role in the regulation of fertility through direct regulation of the GnRH neurons. However, the relative contributions of the two functionally distinct kisspeptin neuron subpopulations to this critical regulation are not fully understood. Here we analyzed the specific projection patterns of kisspeptin neurons originating from either the rostral periventricular nucleus of the third ventricle (RP3V) or the arcuate nucleus (ARN) using a cell-specific, viral-mediated tract-tracing approach. We stereotaxically injected a Cre-dependent recombinant adenovirus encoding farnesylated enhanced green fluorescent protein into the ARN or RP3V of adult male and female mice expressing Cre recombinase in kisspeptin neurons. Fibers from ARN kisspeptin neurons projected widely; however, we did not find any evidence for direct contact with GnRH neuron somata or proximal dendrites in either sex. In contrast, we identified RP3V kisspeptin fibers in close contact with GnRH neuron somata and dendrites in both sexes. Fibers originating from both the RP3V and ARN were observed in close contact with distal GnRH neuron processes in the ARN and in the lateral and internal aspects of the median eminence. Furthermore, GnRH nerve terminals were found in close contact with the proximal dendrites of ARN kisspeptin neurons in the ARN, and ARN kisspeptin fibers were found contacting RP3V kisspeptin neurons in both sexes. Together these data delineate selective zones of kisspeptin neuron inputs to GnRH neurons and demonstrate complex interconnections between the distinct kisspeptin populations and GnRH neurons.
Collapse
Affiliation(s)
- Siew Hoong Yip
- Centre for Neuroendocrinology and Department of Physiology, School of Medical Sciences, University of Otago, Dunedin, New Zealand 9054
| | - Ulrich Boehm
- Centre for Neuroendocrinology and Department of Physiology, School of Medical Sciences, University of Otago, Dunedin, New Zealand 9054
| | - Allan E Herbison
- Centre for Neuroendocrinology and Department of Physiology, School of Medical Sciences, University of Otago, Dunedin, New Zealand 9054
| | - Rebecca E Campbell
- Centre for Neuroendocrinology and Department of Physiology, School of Medical Sciences, University of Otago, Dunedin, New Zealand 9054
| |
Collapse
|
46
|
Yin W, Sun Z, Mendenhall JM, Walker DM, Riha PD, Bezner KS, Gore AC. Expression of Vesicular Glutamate Transporter 2 (vGluT2) on Large Dense-Core Vesicles within GnRH Neuroterminals of Aging Female Rats. PLoS One 2015; 10:e0129633. [PMID: 26053743 PMCID: PMC4459826 DOI: 10.1371/journal.pone.0129633] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 05/10/2015] [Indexed: 11/20/2022] Open
Abstract
The pulsatile release of GnRH is crucial for normal reproductive physiology across the life cycle, a process that is regulated by hypothalamic neurotransmitters. GnRH terminals co-express the vesicular glutamate transporter 2 (vGluT2) as a marker of a glutamatergic phenotype. The current study sought to elucidate the relationship between glutamate and GnRH nerve terminals in the median eminence—the site of GnRH release into the portal capillary vasculature. We also determined whether this co-expression may change during reproductive senescence, and if steroid hormones, which affect responsiveness of GnRH neurons to glutamate, may alter the co-expression pattern. Female Sprague-Dawley rats were ovariectomized at young adult, middle-aged and old ages (~4, 11, and 22 months, respectively) and treated four weeks later with sequential vehicle + vehicle (VEH + VEH), estradiol + vehicle (E2 + VEH), or estradiol + progesterone (E2+P4). Rats were perfused 24 hours after the second hormone treatment. Confocal microscopy was used to determine colocalization of GnRH and vGluT2 immunofluorescence in the median eminence. Post-embedding immunogold labeling of GnRH and vGluT2, and a serial electron microscopy (EM) technique were used to determine the cellular interaction between GnRH terminals and glutamate signaling. Confocal analysis showed that GnRH and vGluT2 immunofluorescent puncta were extensively colocalized in the median eminence and that their density declined with age but was unaffected by short-term hormone treatment. EM results showed that vGluT2 immunoreactivity was extensively associated with large dense-core vesicles, suggesting a unique glutamatergic signaling pathway in GnRH terminals. Our results provide novel subcellular information about the intimate relationship between GnRH terminals and glutamate in the median eminence.
Collapse
Affiliation(s)
- Weiling Yin
- Division of Pharmacology and Toxicology, College of Pharmacy, University of Texas at Austin, Austin, Texas, United States of America
| | - Zengrong Sun
- School of Public Health, Tianjin Medical University, Tianjin, China
| | - John M. Mendenhall
- Institute for Neuroscience, University of Texas at Austin, Austin, Texas, United States of America
| | - Deena M. Walker
- Institute for Neuroscience, University of Texas at Austin, Austin, Texas, United States of America
| | - Penny D. Riha
- Division of Pharmacology and Toxicology, College of Pharmacy, University of Texas at Austin, Austin, Texas, United States of America
| | - Kelsey S. Bezner
- Division of Pharmacology and Toxicology, College of Pharmacy, University of Texas at Austin, Austin, Texas, United States of America
| | - Andrea C. Gore
- Division of Pharmacology and Toxicology, College of Pharmacy, University of Texas at Austin, Austin, Texas, United States of America
- Institute for Neuroscience, University of Texas at Austin, Austin, Texas, United States of America
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, Texas, United States of America
- * E-mail:
| |
Collapse
|
47
|
Clarke H, Dhillo WS, Jayasena CN. Comprehensive Review on Kisspeptin and Its Role in Reproductive Disorders. Endocrinol Metab (Seoul) 2015; 30:124-41. [PMID: 26194072 PMCID: PMC4508256 DOI: 10.3803/enm.2015.30.2.124] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 01/05/2015] [Accepted: 01/12/2015] [Indexed: 02/05/2023] Open
Abstract
Kisspeptin has recently emerged as a key regulator of the mammalian reproductive axis. It is known that kisspeptin, acting centrally via the kisspeptin receptor, stimulates secretion of gonadotrophin releasing hormone (GnRH). Loss of kisspeptin signaling causes hypogonadotrophic hypogonadism in humans and other mammals. Kisspeptin interacts with other neuropeptides such as neurokinin B and dynorphin, to regulate GnRH pulse generation. In addition, a growing body of evidence suggests that kisspeptin signaling be regulated by nutritional status and stress. Kisspeptin may also represent a novel potential therapeutic target in the treatment of fertility disorders. Early human studies suggest that peripheral exogenous kisspeptin administration stimulates gonadotrophin release in healthy adults and in patients with certain forms of infertility. This review aims to concisely summarize what is known about kisspeptin as a regulator of reproductive function, and provide an update on recent advances within this field.
Collapse
Affiliation(s)
- Holly Clarke
- Department of Investigative Medicine, Hammersmith Hospital, Imperial College London, London, UK
| | - Waljit S Dhillo
- Department of Investigative Medicine, Hammersmith Hospital, Imperial College London, London, UK
| | - Channa N Jayasena
- Department of Investigative Medicine, Hammersmith Hospital, Imperial College London, London, UK.
| |
Collapse
|
48
|
Iremonger KJ, Herbison AE. Multitasking in Gonadotropin-Releasing Hormone Neuron Dendrites. Neuroendocrinology 2015; 102:1-7. [PMID: 25300776 DOI: 10.1159/000368364] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 09/10/2014] [Indexed: 11/19/2022]
Abstract
Gonadotropin-releasing hormone (GnRH) neurons integrate synaptic information in their dendrites in order to precisely control GnRH secretion and hence fertility. Recent discoveries concerning the structure and function of GnRH neuron dendrites have shed new light on the control of GnRH neuron excitability and GnRH secretion. This work suggests that GnRH neurons have a unique projection to the median eminence that possesses both dendritic and axonal properties. We propose that this 'dendron' projection allows GnRH neurons to multitask and integrate information in ways that would not be possible in a classically envisioned axon projection.
Collapse
Affiliation(s)
- Karl J Iremonger
- Centre for Neuroendocrinology, Department of Physiology, University of Otago School of Medical Sciences, Dunedin, New Zealand
| | | |
Collapse
|
49
|
Piet R, de Croft S, Liu X, Herbison AE. Electrical properties of kisspeptin neurons and their regulation of GnRH neurons. Front Neuroendocrinol 2015; 36:15-27. [PMID: 24907402 DOI: 10.1016/j.yfrne.2014.05.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 04/01/2014] [Accepted: 05/27/2014] [Indexed: 01/19/2023]
Abstract
Kisspeptin neurons are critical components of the neuronal network controlling the activity of the gonadotropin-releasing hormone (GnRH) neurons. A variety of genetically-manipulated mouse models have recently facilitated the study of the electrical activity of the two principal kisspeptin neuron populations located in the rostral periventricular area of the third ventricle (RP3V) and arcuate nucleus (ARN) in acute brain slices. We discuss here the mechanisms and pathways through which kisspeptin neurons regulate GnRH neuron activity. We then examine the different kisspeptin-green fluorescent protein mouse models being used for kisspeptin electrophysiology and the data obtained to date for RP3V and ARN kisspeptin neurons. In light of these new observations on the spontaneous firing rates, intrinsic membrane properties, and neurotransmitter regulation of kisspeptin neurons, we speculate on the physiological roles of the different kisspeptin populations.
Collapse
Affiliation(s)
- Richard Piet
- Centre for Neuroendocrinology and Department of Physiology, Otago School of Medical Sciences, University of Otago, PO Box 56, Dunedin 9054, New Zealand
| | - Simon de Croft
- Centre for Neuroendocrinology and Department of Physiology, Otago School of Medical Sciences, University of Otago, PO Box 56, Dunedin 9054, New Zealand
| | - Xinhuai Liu
- Centre for Neuroendocrinology and Department of Physiology, Otago School of Medical Sciences, University of Otago, PO Box 56, Dunedin 9054, New Zealand
| | - Allan E Herbison
- Centre for Neuroendocrinology and Department of Physiology, Otago School of Medical Sciences, University of Otago, PO Box 56, Dunedin 9054, New Zealand.
| |
Collapse
|
50
|
Chen X, Sneyd J. A Computational Model of the Dendron of the GnRH Neuron. Bull Math Biol 2014; 77:904-26. [PMID: 25503424 DOI: 10.1007/s11538-014-0052-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 12/03/2014] [Indexed: 10/24/2022]
Abstract
Gonadotropin-releasing hormone (GnRH) neurons have two major processes that have properties of both dendrites (they receive synaptic input from other neurons) and axons (they actively propagate action potentials to the synaptic terminal). These processes have thus been termed dendrons. We construct a stochastic spatiotemporal model of the dendron of the GnRH neuron, with the goal of studying how stochastic synaptic input along the length of the dendron affects the initiation and propagation of action potentials. We show (1) that synaptic inputs closer to the soma are effective controllers of action potential initiation and electrical bursting and (2) that although the effects on the amplitude and width of propagating action potentials are critically dependent on the timing and location of synaptic input addition, the effects remain small. We conclude that although stochastic synaptic input along the length of the dendron is likely to be a major determinant of action potential initiation, it is an unlikely mechanism for controlling whether or not action potentials reach the synaptic terminal. Thus, the role of synaptic inputs situated along the dendron a long way from the site of action potential initiation remains unclear. We also show that the actions of kisspeptin can result in significant modulation of the amount of calcium released by an action potential at the synaptic terminal. Furthermore, we show that the actions of kisspeptin are greatest when multiple effects operate together and that a kisspeptin-induced increase in firing rate is, by itself, less effective at increasing Ca2+ release than is a combination of an increased firing rate, an increase in Ca2+ influx, and an increase in inositol trisphosphate (IP3) production. We conclude that the inherent synergies in the various actions of kisspeptin make it a likely candidate for the precise control of Ca2+ transients at the synaptic terminal.
Collapse
Affiliation(s)
- Xingjiang Chen
- Department of Mathematics, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand,
| | | |
Collapse
|