1
|
Gombert-Labedens M, Vesterdorf K, Fuller A, Maloney SK, Baker FC. Effects of menopause on temperature regulation. Temperature (Austin) 2025; 12:92-132. [PMID: 40330614 PMCID: PMC12051537 DOI: 10.1080/23328940.2025.2484499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 05/08/2025] Open
Abstract
Changes in thermoregulation, notably the emergence of hot flashes, occur during the menopause transition in association with reproductive hormonal changes. Hot flashes constitute the most characteristic symptom of menopause (prevalence of 50-80%), and have a substantial negative effect on quality of life. Here, we review the endocrine changes associated with menopause and the thermoregulatory system and its sensitivity to female sex hormones. We then review current knowledge on the underlying neural mechanisms of hot flashes and how the reproductive and thermoregulatory systems interact in females. We consider the kisspeptin-neurokinin B-dynorphin (KNDy) neuron complex, which becomes hyperactive when estradiol levels decrease. KNDy neurons project from the arcuate nucleus to thermoregulatory areas within the hypothalamic preoptic area, where heat loss mechanisms are triggered, including cutaneous vasodilation and sweating - characteristics of the hot flash. We describe the physiology and measurement of hot flashes and discuss the mixed research findings about thresholds for sweating in symptomatic individuals. We consider the unique situation of hot flashes that arise during sleep, and discuss the relationships between the environment, exercise, and body mass index with hot flashes. We also discuss the unique situation of surgical menopause (with oophorectomy) and cancer therapy, conditions that are associated with frequent, severe, hot flashes. We then provide an overview of treatments of hot flashes, including hormone therapy and targeted neurokinin B-antagonists, recently developed to target the neural mechanism of hot flashes. Finally, we highlight gaps in knowledge about menopausal thermoregulation and hot flashes and suggest future directions for research.
Collapse
Affiliation(s)
| | - Kristine Vesterdorf
- School of Human Sciences, The University of Western Australia, Perth, Australia
| | - Andrea Fuller
- Brain Function Research Group, School of Physiology, University of the Witwatersrand, Johannesburg, South Africa
| | - Shane K. Maloney
- School of Human Sciences, The University of Western Australia, Perth, Australia
- Brain Function Research Group, School of Physiology, University of the Witwatersrand, Johannesburg, South Africa
| | - Fiona C. Baker
- Center for Health Sciences, SRI International, Menlo Park, CA, USA
- Brain Function Research Group, School of Physiology, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
2
|
Koysombat K, Tsoutsouki J, Patel AH, Comninos AN, Dhillo WS, Abbara A. Kisspeptin and neurokinin B: roles in reproductive health. Physiol Rev 2025; 105:707-764. [PMID: 39813600 DOI: 10.1152/physrev.00015.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 10/17/2024] [Accepted: 11/13/2024] [Indexed: 01/18/2025] Open
Abstract
Kisspeptin and neurokinin B (NKB) play a key role in several physiological processes including in puberty, adult reproductive function including the menstrual cycle, as well as mediating the symptoms of menopause. Infundibular kisspeptin neurons, which coexpress NKB, regulate the activity of gonadotropin-releasing hormone (GnRH) neurons and thus the physiological pulsatile secretion of GnRH from the hypothalamus. Outside of their hypothalamic reproductive roles, these peptides are implicated in several physiological functions including sexual behavior and attraction, placental function, and bone health. Over the last two decades, research findings have considerably enhanced our understanding of the physiological regulation of the hypothalamic-pituitary-gonadal (HPG) axis and identified potential therapeutic applications. For example, recognition of the role of kisspeptin as the natural inductor of ovulation has led to research investigating its use as a safer, more physiological trigger of oocyte maturation in in vitro fertilization (IVF) treatment. Moreover, the key role of NKB in the pathophysiology of menopausal hot flashes has led to the development of pharmacological antagonism of this pathway. Indeed, fezolinetant, a neurokinin 3 receptor antagonist, has recently received Food and Drug Administration (FDA) approval for clinical use to treat menopausal vasomotor symptoms. Here, we discuss the roles of kisspeptin and NKB in human physiology, including in the regulation of puberty, menstrual cyclicity, reproductive behavior, pregnancy, menopause, and bone homeostasis. We describe how perturbations of these key physiological processes can result in disease states and consider how kisspeptin and NKB could be exploited diagnostically as well as therapeutically to treat reproductive disorders.
Collapse
Affiliation(s)
- Kanyada Koysombat
- Department of Investigative Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Jovanna Tsoutsouki
- Department of Investigative Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Aaran H Patel
- Department of Investigative Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Alexander N Comninos
- Department of Investigative Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Waljit S Dhillo
- Department of Investigative Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Ali Abbara
- Department of Investigative Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, United Kingdom
| |
Collapse
|
3
|
Sassarini J, Skorupskaite K, McLean J, Lumsden MA, Anderson RA. NK3 receptor antagonist alters the centrally controlled perception of menopausal flushing - a pilot study. Climacteric 2025:1-8. [PMID: 40085748 DOI: 10.1080/13697137.2025.2470462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/29/2025] [Accepted: 02/06/2025] [Indexed: 03/16/2025]
Abstract
OBJECTIVE Kisspeptin/neurokinin/dynorphin (KNDy) signaling links reproductive and thermoregulatory systems, and improvements in menopausal flushing are reported with neurokinin 3 receptor (NK3R) antagonists. A rise in brainstem activity preceding a flush has been proposed as its functional origin, with subsequent activity in the insula and prefrontal cortices reflecting individual perception. Using functional magnetic resonance imaging (fMRI), this study investigated the central effect of the NK3R antagonist MLE4901 during a flush, particularly functional connectivity changes in the salience network. METHOD Five postmenopausal women with flushes completed a 1-week flush diary prior to baseline fMRI, during which hot flushes were triggered by heating. Diaries were continued during 7 days of treatment with the NK3R antagonist MLE4901, with repeat fMRI on day 7. Sternal monitors recorded objective flushing before each fMRI. Connectivity changes in the salience network post flush were assessed. RESULTS Treatment with MLE4901 reduced the subjective flush frequency (from median 6.9 to 1.1 per day; p = 0.02) without changes in objective flushes. Treatment decreased right anterior insula connectivity, which correlated significantly with decreased subjective flushing. CONCLUSION This pilot study demonstrates decreased connectivity in the salience network during NK3R antagonist treatment. This may indicate areas of interest for further targeted fMRI studies and mechanistic investigation of this novel treatment for flushing.
Collapse
Affiliation(s)
- Jenifer Sassarini
- College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | | | - John McLean
- Department of Clinical Physics and Bioengineering (DCPB), NHS Greater Glasgow and Clyde, Glasgow, UK
| | - Mary Ann Lumsden
- College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | | |
Collapse
|
4
|
Szentkirályi-Tóth S, Göcz B, Takács S, Sárvári M, Farkas I, Skrapits K, Rumpler É, Póliska S, Rácz G, Matolcsy A, Ternier G, Fernandois D, Giacobini P, Prévot V, Colledge WH, Wittmann G, Kádár A, Mohácsik P, Gereben B, Fekete C, Hrabovszky E. Estrogen-Regulated Lateral Septal Kisspeptin Neurons Abundantly Project to GnRH Neurons and the Hypothalamic Supramammillary Nucleus. J Neurosci 2025; 45:e1307242024. [PMID: 39746822 PMCID: PMC11841763 DOI: 10.1523/jneurosci.1307-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/04/2025] Open
Abstract
While hypothalamic kisspeptin (KP) neurons play well-established roles in the estrogen-dependent regulation of reproduction, little is known about extrahypothalamic KP-producing (KPLS) neurons of the lateral septum. As established previously, Kiss1 expression in this region is low and regulated by estrogen receptor- and GABAB receptor-dependent mechanisms. Our present experiments on Kiss1-Cre/ZsGreen knock-in mice revealed that transgene expression in the LS begins at Postnatal Day (P)33-36 in females and P40-45 in males and is stimulated by estrogen receptor signaling. Fluorescent cell numbers continue to increase in adulthood and are higher in females. Viral tracing uncovered that the bulk of KPLS fibers joins the medial forebrain bundle and terminates in the hypothalamic supramammillary nucleus. Smaller subsets innervate the medial amygdala or project to other limbic structures. One-quarter of gonadotropin-releasing hormone (GnRH)-immunoreactive perikarya in the preoptic area and their dendrites receive appositions from KPLS axons. OVX adult Kiss1-Cre/ZsGreen mice treated for 4 d with 17β-estradiol or vehicle were used for RNA sequencing studies of laser-microdissected KPLS neurons. The transcriptome included markers of GABAergic and neuropeptidergic (Penk, Cartpt, Vgf) cotransmission and 571 estrogen-regulated transcripts. Estrogen treatment upregulated the acetylcholine receptor transcript Chrm2 and, in slice electrophysiology experiments, caused enhanced muscarinic inhibition of KPLS neurons. Finally, we provided immunohistochemical evidence for homologous neurons in the postmortem human brain, suggesting that KPLS neurons may contribute to evolutionarily conserved regulatory mechanisms. Future studies will need to investigate the putative roles of KPLS neurons in the estrogen-dependent control of GnRH neurons and/or various hypothalamic/limbic functions.
Collapse
Affiliation(s)
- Soma Szentkirályi-Tóth
- Laboratory of Reproductive Neurobiology, HUN-REN Institute of Experimental Medicine, Budapest 1083, Hungary
| | - Balázs Göcz
- Laboratory of Reproductive Neurobiology, HUN-REN Institute of Experimental Medicine, Budapest 1083, Hungary
| | - Szabolcs Takács
- Laboratory of Reproductive Neurobiology, HUN-REN Institute of Experimental Medicine, Budapest 1083, Hungary
| | - Miklós Sárvári
- Laboratory of Reproductive Neurobiology, HUN-REN Institute of Experimental Medicine, Budapest 1083, Hungary
| | - Imre Farkas
- Laboratory of Reproductive Neurobiology, HUN-REN Institute of Experimental Medicine, Budapest 1083, Hungary
| | - Katalin Skrapits
- Laboratory of Reproductive Neurobiology, HUN-REN Institute of Experimental Medicine, Budapest 1083, Hungary
| | - Éva Rumpler
- Laboratory of Reproductive Neurobiology, HUN-REN Institute of Experimental Medicine, Budapest 1083, Hungary
| | - Szilárd Póliska
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen 4032, Hungary
| | - Gergely Rácz
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest 1083, Hungary
| | - András Matolcsy
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest 1083, Hungary
| | - Gaëtan Ternier
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille F-59000, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Lille F-59000, France
| | - Daniela Fernandois
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille F-59000, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Lille F-59000, France
| | - Paolo Giacobini
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille F-59000, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Lille F-59000, France
| | - Vincent Prévot
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille F-59000, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Lille F-59000, France
| | - William H Colledge
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, United Kingdom
| | - Gábor Wittmann
- Laboratory of Integrative Neuroendocrinology, HUN-REN Institute of Experimental Medicine, Budapest 1083, Hungary
| | - Andrea Kádár
- Laboratory of Integrative Neuroendocrinology, HUN-REN Institute of Experimental Medicine, Budapest 1083, Hungary
| | - Petra Mohácsik
- Laboratory of Molecular Cell Metabolism, HUN-REN Institute of Experimental Medicine, Budapest 1083, Hungary
| | - Balázs Gereben
- Laboratory of Molecular Cell Metabolism, HUN-REN Institute of Experimental Medicine, Budapest 1083, Hungary
| | - Csaba Fekete
- Laboratory of Integrative Neuroendocrinology, HUN-REN Institute of Experimental Medicine, Budapest 1083, Hungary
| | - Erik Hrabovszky
- Laboratory of Reproductive Neurobiology, HUN-REN Institute of Experimental Medicine, Budapest 1083, Hungary
| |
Collapse
|
5
|
Zhang W, Jia J, Yang Y, Ye D, Li Y, Li D, Wang J. Estradiol metabolism by gut microbiota in women's depression pathogenesis: inspiration from nature. Front Psychiatry 2025; 16:1505991. [PMID: 39935532 PMCID: PMC11811108 DOI: 10.3389/fpsyt.2025.1505991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 01/10/2025] [Indexed: 02/13/2025] Open
Abstract
The recurrence and treatment resistance of depression remain significant issues, primarily due to an inadequate understanding of its pathogenesis. Recent scientific evidence indicates that gut microbiota influence estradiol metabolism and are associated with the development of depression in nonpremenopausal women. Integrating existing studies on the regulation of estradiol metabolism by microorganisms in nature and the relevance of its degradation products to depression, recent scientific explorations have further elucidated the key mechanisms by which gut microbiota catabolize estradiol through specific metabolic pathways. These emerging scientific findings suggest that the unique metabolic effects of gut microbiota on estradiol may be one of the central drivers in the onset and course of depression in non-menopausal women.
Collapse
Affiliation(s)
- Wei Zhang
- Division of Colorectal Surgery, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Taiyuan, China
| | - Jinghan Jia
- Division of Colorectal Surgery, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Taiyuan, China
| | - Yuhang Yang
- Division of Colorectal Surgery, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Taiyuan, China
| | - Dawei Ye
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yan Li
- Neurology, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Taiyuan, China
| | - Di Li
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinxi Wang
- Division of Colorectal Surgery, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
6
|
Fidecicchi T, Giannini A, Chedraui P, Luisi S, Battipaglia C, Genazzani AR, Genazzani AD, Simoncini T. Neuroendocrine mechanisms of mood disorders during menopause transition: A narrative review and future perspectives. Maturitas 2024; 188:108087. [PMID: 39111089 DOI: 10.1016/j.maturitas.2024.108087] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 07/29/2024] [Accepted: 07/31/2024] [Indexed: 09/01/2024]
Abstract
The menopause transition is an important period in a woman's life, during which she is at an increased risk of mood disorders. Estrogen and progesterone fluctuations during the menopausal transition and very low levels of estradiol after menopause have a profound effect on the central nervous system (CNS), causing an imbalance between excitatory and inhibitory inputs. Changes in neurotransmission and neuronal interactions that occur with estradiol withdrawal disrupt the normal neurological balance and may be associated with menopausal symptoms. Hot flushes, depressed mood and anxiety are all symptoms of menopause that are a consequence of the complex changes that occur in the CNS, involving many signaling pathways and neurotransmitters (i.e. γ-aminobutyric acid, serotonin, dopamine), neurosteroids (i.e. allopregnanolone), and neuropeptides (i.e. kisspeptin, neurokinin B). All these pathways are closely linked, and the complex interactions that exist are not yet fully understood. This review summarizes the neuroendocrine changes in the CNS during the menopausal transition, with particular emphasis on those that underlie mood changes.
Collapse
Affiliation(s)
- Tiziana Fidecicchi
- Department of Clinical and Experimental Medicine, Division of Gynecology and Obstetrics, University of Pisa, Via Roma 67, 56126 Pisa, PI, Italy
| | - Andrea Giannini
- Department of Clinical and Experimental Medicine, Division of Gynecology and Obstetrics, University of Pisa, Via Roma 67, 56126 Pisa, PI, Italy.
| | - Peter Chedraui
- Escuela de Posgrado en Salud, Universidad Espíritu Santo, Av. Samborondón 5, Samborondón 092301, Ecuador
| | - Stefano Luisi
- Department of Clinical and Experimental Medicine, Division of Gynecology and Obstetrics, University of Pisa, Via Roma 67, 56126 Pisa, PI, Italy.
| | - Christian Battipaglia
- Gynecological Endocrinology Center, Department of Obstetrics and Gynecology, University of Modena and Reggio Emilia, Largo del Pozzo, 71, 41125 Modena, MO, Italy.
| | - Andrea R Genazzani
- Department of Clinical and Experimental Medicine, Division of Gynecology and Obstetrics, University of Pisa, Via Roma 67, 56126 Pisa, PI, Italy
| | - Alessandro D Genazzani
- Gynecological Endocrinology Center, Department of Obstetrics and Gynecology, University of Modena and Reggio Emilia, Largo del Pozzo, 71, 41125 Modena, MO, Italy
| | - Tommaso Simoncini
- Department of Clinical and Experimental Medicine, Division of Gynecology and Obstetrics, University of Pisa, Via Roma 67, 56126 Pisa, PI, Italy.
| |
Collapse
|
7
|
Hrabovszky E. An historical step in our understanding of hypothalamic oestrogen feedback. Nat Rev Endocrinol 2024; 20:258. [PMID: 38418670 DOI: 10.1038/s41574-024-00968-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Affiliation(s)
- Erik Hrabovszky
- Reproductive Neurobiology Research Group, Institute of Experimental Medicine, Budapest, Hungary.
| |
Collapse
|
8
|
Koysombat K, McGown P, Nyunt S, Abbara A, Dhillo WS. New advances in menopause symptom management. Best Pract Res Clin Endocrinol Metab 2024; 38:101774. [PMID: 37076317 DOI: 10.1016/j.beem.2023.101774] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/21/2023]
Abstract
Vasomotor symptoms (VMS) are characteristic of menopause experienced by over 75% of postmenopausal women with significant health and socioeconomic implications. Although the average duration of symptoms is seven years, 10% of women experience symptoms for more than a decade. Although menopausal hormone therapy (MHT) remains an efficacious and cost-effective treatment, its use may not be suitable in all women, such as those at an increased risk of breast cancer or gynaecological malignancy. The neurokinin B (NKB) signaling pathway, together with its intricate connection to the median preoptic nucleus (MnPO), has been postulated to provide integrated reproductive and thermoregulatory responses, with a central role in mediating postmenopausal VMS. This review describes the physiological hypothalamo-pituitary-ovary (HPO) axis, and subsequently the neuroendocrine changes that occur with menopause using evidence derived from animal and human studies. Finally, data from the latest clinical trials using novel therapeutic agents that antagonise NKB signaling are reviewed.
Collapse
Affiliation(s)
- Kanyada Koysombat
- Section of Investigative Medicine, Imperial College London, London, United Kingdom; Department of Endocrinology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Patrick McGown
- Section of Investigative Medicine, Imperial College London, London, United Kingdom
| | - Sandhi Nyunt
- Section of Investigative Medicine, Imperial College London, London, United Kingdom; Department of Endocrinology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Ali Abbara
- Section of Investigative Medicine, Imperial College London, London, United Kingdom; Department of Endocrinology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Waljit S Dhillo
- Section of Investigative Medicine, Imperial College London, London, United Kingdom; Department of Endocrinology, Imperial College Healthcare NHS Trust, London, United Kingdom.
| |
Collapse
|
9
|
Barlow DH. Continuing progress on vasomotor symptoms. Menopause 2023; 30:235-236. [PMID: 36749902 DOI: 10.1097/gme.0000000000002160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
|
10
|
Simon JA, Anderson RA, Ballantyne E, Bolognese J, Caetano C, Joffe H, Kerr M, Panay N, Seitz C, Seymore S, Trower M, Zuurman L, Pawsey S. Efficacy and safety of elinzanetant, a selective neurokinin-1,3 receptor antagonist for vasomotor symptoms: a dose-finding clinical trial (SWITCH-1). Menopause 2023; 30:239-246. [PMID: 36720081 PMCID: PMC9970022 DOI: 10.1097/gme.0000000000002138] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/26/2022] [Accepted: 10/26/2022] [Indexed: 02/01/2023]
Abstract
OBJECTIVE Neurokinin (NK)-3 and NK-1 receptors have been implicated in the etiology of vasomotor symptoms (VMS) and sleep disturbances associated with menopause. This phase 2b, adaptive, dose-range finding study aimed to assess the efficacy and safety of multiple doses of elinzanetant (NT-814), a selective NK-1,3 receptor antagonist, in women experiencing VMS associated with menopause, and investigate the impact of elinzanetant on sleep and quality of life. METHODS Postmenopausal women aged 40 to 65 years who experienced seven or more moderate-to-severe VMS per day were randomized to receive elinzanetant 40, 80, 120, or 160 mg or placebo once daily using an adaptive design algorithm. Coprimary endpoints were reduction in mean frequency and severity of moderate-to-severe VMS at weeks 4 and 12. Secondary endpoints included patient-reported assessments of sleep and quality of life. RESULTS Elinzanetant 120 mg and 160 mg achieved reductions in VMS frequency versus placebo from week 1 throughout 12 weeks of treatment. Least square mean reductions were statistically significant versus placebo at both primary endpoint time points for elinzanetant 120 mg (week 4: -3.93 [SE, 1.02], P < 0.001; week 12: -2.95 [1.15], P = 0.01) and at week 4 for elinzanetant 160 mg (-2.63 [1.03]; P = 0.01). Both doses also led to clinically meaningful improvements in measures of sleep and quality of life. All doses of elinzanetant were well tolerated. CONCLUSIONS Elinzanetant is an effective and well-tolerated nonhormone treatment option for postmenopausal women with VMS and associated sleep disturbance. Elinzanetant also improves quality of life in women with VMS.
Collapse
Affiliation(s)
- James A. Simon
- From the George Washington University, IntimMedicine Specialists, Washington, DC
| | - Richard A. Anderson
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | | | | | | | - Hadine Joffe
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Mary Kerr
- NeRRe Therapeutics, Stevenage, United Kingdom
| | - Nick Panay
- Queen Charlotte's and Chelsea and Westminster Hospitals, Imperial College, London, United Kingdom
| | | | | | - Mike Trower
- NeRRe Therapeutics, Stevenage, United Kingdom
| | | | | |
Collapse
|
11
|
Stress, kisspeptin, and functional hypothalamic amenorrhea. Curr Opin Pharmacol 2022; 67:102288. [PMID: 36103784 DOI: 10.1016/j.coph.2022.102288] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/04/2022] [Accepted: 08/04/2022] [Indexed: 01/25/2023]
Abstract
Functional hypothalamic amenorrhea (FHA) is the most common cause of secondary amenorrhea in women of reproductive age. FHA is predominantly caused by stress, decreased caloric intake, excessive exercise, or a combination thereof. These physical, psychological, and metabolic stressors cause aberration in the pulsatile release of gonadotropin-releasing hormone (GnRH) and subsequently impair function of the hypothalamic-pituitary-ovarian (HPO) axis. Various neurotransmitters acting in the central nervous system are involved in control of the HPO axis and of these, kisspeptin is one of the most important. Corticotropin-releasing hormone (CRH), also inhibits the pulsatile secretion of GnRH and also acts as an intermediary between stress factors and the reproductive system. One of the main ongoing concerns in patients with FHA is chronic hypoestrogenism, a condition, which is associated with sexual dysfunction and infertility. It may also lead to osteoporosis, and predispose to neurodegenerative and cardiovascular diseases. Treatment of FHA requires the elimination of causative factors, however, making the necessary lifestyle changes is not always easy to initiate and maintain. Broadening our knowledge of the complex neural mechanisms regulating reproductive function in which kisspeptin plays a key role can help in the development of new treatment options such as the potential of kisspeptin receptor agonists for patients with FHA.
Collapse
|
12
|
Transcriptome profiling of kisspeptin neurons from the mouse arcuate nucleus reveals new mechanisms in estrogenic control of fertility. Proc Natl Acad Sci U S A 2022; 119:e2113749119. [PMID: 35763574 PMCID: PMC9271166 DOI: 10.1073/pnas.2113749119] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Kisspeptin neurons in the mediobasal hypothalamus (MBH) are critical targets of ovarian estrogen feedback regulating mammalian fertility. To reveal molecular mechanisms underlying this signaling, we thoroughly characterized the estrogen-regulated transcriptome of kisspeptin cells from ovariectomized transgenic mice substituted with 17β-estradiol or vehicle. MBH kisspeptin neurons were harvested using laser-capture microdissection, pooled, and subjected to RNA sequencing. Estrogen treatment significantly (p.adj. < 0.05) up-regulated 1,190 and down-regulated 1,139 transcripts, including transcription factors, neuropeptides, ribosomal and mitochondrial proteins, ion channels, transporters, receptors, and regulatory RNAs. Reduced expression of the excitatory serotonin receptor-4 transcript (Htr4) diminished kisspeptin neuron responsiveness to serotonergic stimulation. Many estrogen-regulated transcripts have been implicated in puberty/fertility disorders. Patients (n = 337) with congenital hypogonadotropic hypogonadism (CHH) showed enrichment of rare variants in putative CHH-candidate genes (e.g., LRP1B, CACNA1G, FNDC3A). Comprehensive characterization of the estrogen-dependent kisspeptin neuron transcriptome sheds light on the molecular mechanisms of ovary-brain communication and informs genetic research on human fertility disorders.
Collapse
|
13
|
Kisspeptin Modulation of Reproductive Function. ENDOCRINES 2022. [DOI: 10.3390/endocrines3030029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Kisspeptin is a peptide expressed mainly in the infundibular nucleus of the hypothalamus. Kisspeptin plays a crucial role in the regulation of reproductive functions. It is regarded as the most important factor responsible for the control of the hypothalamic–pituitary–gonadal axis, the onset of puberty, and the regulation of menstruation and fertility. Kisspeptin activity influences numerous processes such as steroidogenesis, follicular maturation, ovulation, and ovarian senescence. The identification of kisspeptin receptor mutations that cause hypogonadotropic hypogonadism has initiated studies on the role of kisspeptin in puberty. Pathologies affecting the neurons secreting kisspeptin play a major role in the development of PCOS, functional hypothalamic amenorrhea, and perimenopausal vasomotor symptoms. Kisspeptin analogs (both agonists and antagonists), therefore, may be beneficial as therapy in those afflicted with such pathologies. The aim of this review is to summarize the influence of kisspeptin in the physiology and pathology of the reproductive system in humans, as well as its potential use in therapy.
Collapse
|
14
|
Nilsson S, Henriksson M, Berin E, Engblom D, Holm ACS, Hammar M. Resistance training reduced luteinising hormone levels in postmenopausal women in a substudy of a randomised controlled clinical trial: A clue to how resistance training reduced vasomotor symptoms. PLoS One 2022; 17:e0267613. [PMID: 35617333 PMCID: PMC9135255 DOI: 10.1371/journal.pone.0267613] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 03/16/2022] [Indexed: 11/22/2022] Open
Abstract
Background Vasomotor symptoms (VMS) are common around menopause. Menopausal hormone therapy is the most effective treatment for VMS. Physical exercise has been proposed as an alternative treatment since physically active women have previously been found to experience fewer VMS than inactive women. In our randomised controlled trial on resistance training to treat VMS, sympoms were reduced by 50% in the intervention group compared with the control group. Objectives To propose a mechanism to explain how resistance training reduced VMS and to assess if luteinizing hormone (LH) and follicle stimulating hormone (FSH) were affected in accordance with the proposed mechanism. Trial design and methods A substudy of a randomized controlled trial on 65 postmenopausal women with VMS and low physical activity who were randomised to 15 weeks of resistance training three times per week (n = 33) or to a control group (n = 32). To be regarded compliant to the intervention we predecided a mean of two training sessions per week. The daily number of VMS were registered before and during the 15 weeks. Blood samples were drawn for analysis of LH and FSH at baseline and after 15 weeks. Results LH decreased significantly in the compliant intervention group compared with the control group (-4.0±10.6 versus 2.9±9.0, p = 0.028 with Mann-Whitney U test). FSH also decreased in the compliant intervention group compared with the control group, however not enough to reach statistical significance (-3.5±16.3 versus 3.2±18.2, p = 0.063 with Mann-Whitney U test). As previously published the number of hot flushes decreased significantly more in the intervention group than in the control group but there was no association between change in LH or FSH and in number of VMS. Conclusions We propose that endogenous opiods such as β-endorphin or dynorphin produced during resistance training decreased VMS by stimulating KNDγ-neurons to release neurokinin B to the hypothalamic thermoregulatory centre. Through effects on KNDγ-neurons, β-endorphin could also inhibit GnRH and thereby decrease the production of LH and FSH. The significanty decreased LH in the compliant intervention group compared with the control group was in accordance with the proposed mechanism.
Collapse
Affiliation(s)
- Sigrid Nilsson
- Obstetrics and Gynaecology, Division of Children’s and Women’s Health, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Moa Henriksson
- Obstetrics and Gynaecology, Division of Children’s and Women’s Health, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Emilia Berin
- Obstetrics and Gynaecology, Division of Children’s and Women’s Health, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - David Engblom
- Division of Cell Biology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Anna-Clara Spetz Holm
- Obstetrics and Gynaecology, Division of Children’s and Women’s Health, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Mats Hammar
- Obstetrics and Gynaecology, Division of Children’s and Women’s Health, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- * E-mail:
| |
Collapse
|
15
|
Campbell RE, Coolen LM, Hoffman GE, Hrabovszky E. Highlights of neuroanatomical discoveries of the mammalian gonadotropin-releasing hormone system. J Neuroendocrinol 2022; 34:e13115. [PMID: 35502534 PMCID: PMC9232911 DOI: 10.1111/jne.13115] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/09/2022] [Accepted: 03/01/2022] [Indexed: 11/17/2022]
Abstract
The anatomy and morphology of gonadotropin-releasing hormone (GnRH) neurons makes them both a joy and a challenge to investigate. They are a highly unique population of neurons given their developmental migration into the brain from the olfactory placode, their relatively small number, their largely scattered distribution within the rostral forebrain, and, in some species, their highly varied individual anatomical characteristics. These unique features have posed technological hurdles to overcome and promoted fertile ground for the establishment and use of creative approaches. Historical and more contemporary discoveries defining GnRH neuron anatomy remain critical in shaping and challenging our views of GnRH neuron function in the regulation of reproductive function. We begin this review with a historical overview of anatomical discoveries and developing methodologies that have shaped our understanding of the reproductive axis. We then highlight significant discoveries across specific groups of mammalian species to address some of the important comparative aspects of GnRH neuroanatomy. Lastly, we touch on unresolved questions and opportunities for future neuroanatomical research on this fascinating and important population of neurons.
Collapse
Affiliation(s)
- Rebecca E. Campbell
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical SciencesUniversity of OtagoDunedinNew Zealand
| | - Lique M. Coolen
- Department of Biological SciencesKent State UniversityKentOhioUSA
| | | | - Erik Hrabovszky
- Laboratory of Reproductive NeurobiologyInstitute of Experimental MedicineBudapestHungary
| |
Collapse
|
16
|
Xie Q, Kang Y, Zhang C, Xie Y, Wang C, Liu J, Yu C, Zhao H, Huang D. The Role of Kisspeptin in the Control of the Hypothalamic-Pituitary-Gonadal Axis and Reproduction. Front Endocrinol (Lausanne) 2022; 13:925206. [PMID: 35837314 PMCID: PMC9273750 DOI: 10.3389/fendo.2022.925206] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 05/30/2022] [Indexed: 01/07/2023] Open
Abstract
The discovery of kisspeptin as a critical central regulatory factor of GnRH release has given people a novel understanding of the neuroendocrine regulation in human reproduction. Kisspeptin activates the signaling pathway by binding to its receptor kisspeptin receptor (KISS1R) to promote GnRH secretion, thereby regulating the hypothalamic-pituitary-gonadal axis (HPG) axis. Recent studies have shown that kisspeptin neurons located in arcuate nucleus (ARC) co-express neurokinin B (NKB) and dynorphin (Dyn). Such neurons are called KNDy neurons. KNDy neurons participate in the positive and negative feedback of estrogen to GnRH secretion. In addition, kisspeptin is a key factor in the initiation of puberty, and also regulates the processes of female follicle development, oocyte maturation, and ovulation through the HPG axis. In male reproduction, kisspeptin also plays an important role, getting involved in the regulation of Leydig cells, spermatogenesis, sperm functions and reproductive behaviors. Mutations in the KISS1 gene or disorders of the kisspeptin/KISS1R system may lead to clinical symptoms such as idiopathic hypogonadotropic hypogonadism (iHH), central precocious puberty (CPP) and female infertility. Understanding the influence of kisspeptin on the reproductive axis and related mechanisms will help the future application of kisspeptin in disease diagnosis and treatment. In this review, we critically appraise the role of kisspeptin in the HPG axis, including its signaling pathways, negative and positive feedback mechanisms, and its control on female and male reproduction.
Collapse
Affiliation(s)
- Qinying Xie
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yafei Kang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chenlu Zhang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ye Xie
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chuxiong Wang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiang Liu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Caiqian Yu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hu Zhao
- Department of Human Anatomy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Donghui Huang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
17
|
Sullivan AE, Tappan SJ, Angstman PJ, Rodriguez A, Thomas GC, Hoppes DM, Abdul-Karim MA, Heal ML, Glaser JR. A Comprehensive, FAIR File Format for Neuroanatomical Structure Modeling. Neuroinformatics 2022; 20:221-240. [PMID: 34601704 PMCID: PMC8975944 DOI: 10.1007/s12021-021-09530-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2021] [Indexed: 01/09/2023]
Abstract
With advances in microscopy and computer science, the technique of digitally reconstructing, modeling, and quantifying microscopic anatomies has become central to many fields of biological research. MBF Bioscience has chosen to openly document their digital reconstruction file format, the Neuromorphological File Specification, available at www.mbfbioscience.com/filespecification (Angstman et al., 2020). The format, created and maintained by MBF Bioscience, is broadly utilized by the neuroscience community. The data format's structure and capabilities have evolved since its inception, with modifications made to keep pace with advancements in microscopy and the scientific questions raised by worldwide experts in the field. More recent modifications to the neuromorphological file format ensure it abides by the Findable, Accessible, Interoperable, and Reusable (FAIR) data principles promoted by the International Neuroinformatics Coordinating Facility (INCF; Wilkinson et al., Scientific Data, 3, 160018,, 2016). The incorporated metadata make it easy to identify and repurpose these data types for downstream applications and investigation. This publication describes key elements of the file format and details their relevant structural advantages in an effort to encourage the reuse of these rich data files for alternative analysis or reproduction of derived conclusions.
Collapse
|
18
|
Tahara A, Takamatsu H, Ohtake A, Tanaka-Amino K, Kaku S. Effects of neurokinin 3 receptor antagonist fezolinetant on hot flash-like symptoms in ovariectomized rats. Eur J Pharmacol 2021; 905:174207. [PMID: 34048742 DOI: 10.1016/j.ejphar.2021.174207] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 05/19/2021] [Accepted: 05/21/2021] [Indexed: 11/16/2022]
Abstract
The majority of women experience vasomotor symptoms (VMS), such as hot flashes and night sweats, during the menopausal transition. Recent evidence strongly suggests a connection between neurokinin 3 (NK3) receptor signaling and VMS associated with menopause. The NK3 receptor antagonist fezolinetant is currently in phase 3 development for treatment of moderate to severe VMS associated with menopause. We investigated the pharmacological effects of repeated administration of fezolinetant on levels of sex hormones and gonadotropins, neuronal activity in the hypothalamus, and skin temperature as an index of hot flash-like symptoms in ovariectomized rats as a model of menopause. Ovariectomized rats exhibited several typical menopausal symptoms: hyperphagia, increased body weight, significantly decreased plasma estradiol levels, increased luteinizing hormone (LH) and follicle-stimulating hormone (FSH) levels, and significantly increased skin temperature. Increased c-Fos expression (an indirect marker of neuronal activity) in median preoptic nucleus (MnPO) hypothalamic neurons was also observed in ovariectomized rats. Repeated oral administration of fezolinetant (1-10 mg/kg, twice daily) for 1 week dose-dependently reduced plasma LH levels without affecting estradiol or FSH levels, inhibited the activation of MnPO neurons, and attenuated hot flash-like symptoms. In addition, fezolinetant dose-dependently reduced hyperphagia and weight gain in ovariectomized rats. These preclinical findings suggest that fezolinetant attenuates hot flash-like symptoms via inhibition of neuronal activity in the MnPO of ovariectomized rats and provides further support for the ongoing clinical development of fezolinetant for the treatment of VMS associated with menopause.
Collapse
Affiliation(s)
- Atsuo Tahara
- Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan.
| | | | - Akiyoshi Ohtake
- Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan
| | | | - Seiji Kaku
- Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan
| |
Collapse
|
19
|
Hypothalamic neurokinin signalling and its application in reproductive medicine. Pharmacol Ther 2021; 230:107960. [PMID: 34273412 DOI: 10.1016/j.pharmthera.2021.107960] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/06/2021] [Accepted: 07/08/2021] [Indexed: 12/29/2022]
Abstract
The discovery of the essential requirement for kisspeptin and subsequently neurokinin B signalling for human reproductive function has sparked renewed interest in the neuroendocrinology of reproduction. A key discovery has been a population of cells co-expressing both these neuropeptides and dynorphin in the hypothalamus, directly regulating gonadotropin hormone releasing hormone (GnRH) secretion and thus pituitary secretion of gonadotropins. These neurons also project to the vasomotor centre, and their overactivity in estrogen deficiency results in the common and debilitating hot flushes of the menopause. Several antagonists to the neurokinin 3 receptor, for which neurokinin B is the endogenous ligand, have been developed, and are entering clinical studies in human reproductive function and clinical trials. Even single doses can elicit marked declines in testosterone levels in men, and their use has elicited evidence of the regulation of ovarian follicle growth in women. The most advanced indication is the treatment of menopausal vasomotor symptoms, where these drugs show remarkable results in both the degree and speed of symptom control. A range of other reproductive indications are starting to be explored, notably in polycystic ovary syndrome, the most common endocrinopathy in women.
Collapse
|
20
|
Depypere H, Lademacher C, Siddiqui E, Fraser GL. Fezolinetant in the treatment of vasomotor symptoms associated with menopause. Expert Opin Investig Drugs 2021; 30:681-694. [PMID: 33724119 DOI: 10.1080/13543784.2021.1893305] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Introduction: Although international clinical practice guidelines recognize a continued role for menopausal hormone therapy (HT), particularly for symptomatic women <60 years of age or within 10 years of menopause, safety and tolerability concerns have discouraged HT use due to potential links with a perceived increased risk of hormone-dependent cancers, and an established risk of stroke and venous thromboembolism. There is therefore a need for safe, effective non-hormonal therapy for relief of menopausal vasomotor symptoms (VMS).Areas covered: This narrative review summarizes the dataset accrued for fezolinetant, a neurokinin-3 receptor (NK3R) antagonist in clinical development for menopause-associated VMS.Expert opinion: Altered signaling in neuroendocrine circuits at menopause leads to VMS wherein NK3R activity plays a key role to modulate the thermoregulatory center in a manner conducive to triggering the 'hot flash' response. Thus, a new generation of NK3R antagonists has entered clinical development to specifically target the mechanistic basis of VMS. Fezolinetant is the most advanced NK3R antagonist in terms of stage of clinical development. Results to date have demonstrated rapid and substantial reduction in VMS frequency and severity and associated improvements in health-related quality of life. NK3R antagonists offer a non-hormonal alternative to HT for the treatment of menopause-related VMS.
Collapse
Affiliation(s)
- Herman Depypere
- Breast and Menopause Clinic, University Hospital, Ghent, Belgium
| | | | - Emad Siddiqui
- Medical Affairs, Astellas Pharma Medical and Development, Chertsey, UK
| | - Graeme L Fraser
- Former Chief Scientific Officer of Ogeda SA, Gosselies, Belgium and Consultant for Astellas Pharma Inc
| |
Collapse
|
21
|
Effects of NT-814, a dual neurokinin 1 and 3 receptor antagonist, on vasomotor symptoms in postmenopausal women: a placebo-controlled, randomized trial. ACTA ACUST UNITED AC 2021; 27:498-505. [PMID: 32068688 PMCID: PMC7188053 DOI: 10.1097/gme.0000000000001500] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
OBJECTIVES To evaluate the safety, pharmacokinetics, and preliminary efficacy of NT-814, a dual neurokinin 1,3 antagonist, in postmenopausal women with vasomotor symptoms (hot flashes). METHODS We completed a double-blind, randomized, placebo-controlled trial in three US clinical research units in 76 postmenopausal women with moderate/severe hot flashes. Participants were randomized to 14 days of once-daily NT-814 or placebo within each of four sequential dose cohorts; 50, 100, 150, and 300 mg. Participants completed diaries of hot flash frequency and severity and waking due to night sweats before (baseline) and during treatment. RESULTS All prespecified efficacy parameters (24-h hot flash frequency and severity, frequency of waking due to night sweats) decreased in all groups (including placebo). Mean reduction from baseline at week 2 in moderate/severe hot flash frequency was 37% in the placebo group and, respectively, 24% (P = 0.048 vs placebo), 59% (P = 0.155), 84% (P < 0.001) and 66% (P = 0.022) in the 50 mg, 100 mg, 150 mg, and 300 mg NT-814 groups; in waking due to night sweats reduction was 20% (P = 0.059), 55% (P = 0.135), 81% (P < 0.001), and 63% (P = 0.031) in the NT-814 groups and 32% in the placebo group. The improvement with NT-814 ≥150 mg was also evident in the first week of treatment. The most common treatment-related adverse events were mild somnolence and headache, more frequently in the 300 mg group. Safety monitoring identified no concerns. CONCLUSIONS Once-daily NT-814 (≥150 mg/d) resulted in a rapid, marked improvement in hot flashes and waking due to night sweats. No safety concerns were identified. Doses up to 300 mg were well tolerated.
Collapse
|
22
|
Bakker J. Kisspeptin and neurokinin B expression in the human hypothalamus: Relation to reproduction and gender identity. HANDBOOK OF CLINICAL NEUROLOGY 2021; 180:297-313. [PMID: 34225936 DOI: 10.1016/b978-0-12-820107-7.00018-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Gonadotropin-releasing hormone (GnRH) neurons in the hypothalamus are at the core of reproductive functioning. GnRH released into the median eminence regulates the secretion of the gonadotropins from the anterior pituitary, which in turn activates gametogenesis and steroid synthesis by the gonads. The GnRH system displays functional sex differences: GnRH is secreted in pulses at a constant frequency in men, whereas in women, pulse frequency varies over the menstrual cycle. In both sexes, GnRH release is regulated by sex steroid hormones, acting at the level of the hypothalamus and the anterior pituitary in a classic feedback loop. Because GnRH neurons do not express sex steroid receptors, hormone effects on GnRH release are presumed to be mediated indirectly through other steroid-sensitive neuronal systems, which then converge onto GnRH cell bodies and/or terminals. Human genetic studies demonstrated that kisspeptin (KP) as well as neurokinin B (NKB) signaling are both potent regulators of GNRH secretion. In humans, postmortem studies using immunohistochemistry have shown that women have higher KP and NKB expression in the infundibular nucleus than men. Sex differences in KP expression are present throughout life, which is from the infant/prepubertal into the elderly period, whereas sex differences in NKB expression do not emerge until adulthood. KP and NKB are often coexpressed together with dynorphin by the same population of neurons, also known as KDNy neurons in other species. Indeed, significant coexpression between KP and NKB but not with Dynorphin has been observed thereby challenging the KDNy concept in humans. Female-typical expression of both KP and NKB were observed in the infundibular nucleus of trans women (male sex assigned at birth and female gender identity). Taken together, sex differences in KP and NKB expression most likely reflect organizational actions of sex steroid hormones on the developing brain but they also remain sensitive to circulating sex steroids in adulthood. The female-dominant sex difference in infundibular KP and NKB expression suggests that this brain region is most likely involved in both the negative and positive feedback actions of estrogens on GnRH secretion. Finally, the sex-reversal observed in KP and NKB expression in trans women might reflect, at least partially, an atypical sexual differentiation of the brain.
Collapse
Affiliation(s)
- Julie Bakker
- GIGA Neurosciences, Liège University, Liège, Belgium.
| |
Collapse
|
23
|
Modi M, Dhillo WS. The neuroendocrinology of the preoptic area in menopause: Symptoms and therapeutic strategies. HANDBOOK OF CLINICAL NEUROLOGY 2021; 179:455-460. [PMID: 34225982 DOI: 10.1016/b978-0-12-819975-6.00029-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The preoptic area of the hypothalamus is the central hub of thermoregulation in mammals, coordinating autonomic heat-effector pathways in response to sensory information from the ambient and internal environment. This aims to maintain temperature homeostasis at a predetermined thermoregulatory set-point. However, hormonal and neuronal changes during the menopause, including estrogen deficiency, disrupt these normal thermoregulatory responses. This results in abnormal activation of heat dissipation effectors, manifesting clinically as hot flush symptoms. Neurokinin B (NKB) signaling via the neurokinin-3 receptor (NK3R) within the preoptic area is thought to play an important role in the pathophysiology of hot flushes. Therefore attenuation of the NKB/NK3R signaling pathway has garnered much interest as a novel therapeutic target for the amelioration of menopausal hot flushes. Recent clinical trials have demonstrated that NK3R antagonists can produce rapid and sustained improvements in hot flush frequency, severity, and quality of life, without the need for estrogen exposure. Therefore NK3R antagonists are fast emerging as a safe and efficacious alternative to hormone replacement therapy, the current gold standard of treatment.
Collapse
Affiliation(s)
- Manish Modi
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, United Kingdom
| | - Waljit Singh Dhillo
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, United Kingdom.
| |
Collapse
|
24
|
Hrabovszky E, Takács S, Rumpler É, Skrapits K. The human hypothalamic kisspeptin system: Functional neuroanatomy and clinical perspectives. HANDBOOK OF CLINICAL NEUROLOGY 2021; 180:275-296. [PMID: 34225935 DOI: 10.1016/b978-0-12-820107-7.00017-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In mammals, kisspeptin neurons are the key components of the hypothalamic neuronal networks that regulate the onset of puberty, account for the pulsatile secretion of gonadotropin-releasing hormone (GnRH) and mediate negative and positive estrogen feedback signals to GnRH neurons. Being directly connected anatomically and functionally to the hypophysiotropic GnRH system, the major kisspeptin cell groups of the preoptic area/rostral hypothalamus and the arcuate (or infundibular) nucleus, respectively, are ideally positioned to serve as key nodes which integrate various types of environmental, endocrine, and metabolic signals that can influence fertility. This chapter provides an overview of the current state of knowledge on the anatomy, functions, and plasticity of brain kisspeptin systems based on the wide literature available from different laboratory and domestic species. Then, the species-specific features of human hypothalamic kisspeptin neurons are described, covering their topography, morphology, unique neuropeptide content, plasticity, and connectivity to hypophysiotropic GnRH neurons. Some newly emerging roles of central kisspeptin signaling in behavior and finally, clinical perspectives, are discussed.
Collapse
Affiliation(s)
- Erik Hrabovszky
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary.
| | - Szabolcs Takács
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Éva Rumpler
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Katalin Skrapits
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| |
Collapse
|
25
|
A phase 2b, randomized, placebo-controlled, double-blind, dose-ranging study of the neurokinin 3 receptor antagonist fezolinetant for vasomotor symptoms associated with menopause. ACTA ACUST UNITED AC 2020; 27:382-392. [PMID: 32102086 PMCID: PMC7147405 DOI: 10.1097/gme.0000000000001510] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Objective: Menopausal vasomotor symptoms (VMS) may result from altered thermoregulatory control in brain regions innervated by neurokinin 3 receptor-expressing neurons. This phase 2b study evaluated seven dosing regimens of fezolinetant, a selective neurokinin 3 receptor antagonist, as a nonhormone approach for the treatment of VMS. Methods: Menopausal women aged >40-65 years with moderate/severe VMS (≥50 episodes/wk) were randomized (double-blind) to fezolinetant 15, 30, 60, 90 mg BID or 30, 60, 120 mg QD, or placebo for 12 weeks. Primary outcomes were reduction in moderate/severe VMS frequency and severity ([number of moderate VMS × 2] + [number of severe VMS × 3]/total daily moderate/severe VMS) at weeks 4 and 12. Response (≥50% reduction in moderate/severe VMS frequency) was a key secondary outcome. Results: Of 352 treated participants, 287 completed the study. Fezolinetant reduced moderate/severe VMS frequency by −1.9 to −3.5/day at week 4 and −1.8 to −2.6/day at week 12 (all P < 0.05 vs placebo). Mean difference from placebo in VMS severity score was −0.4 to −1 at week 4 (all doses P < 0.05) and −0.2 to −0.6 at week 12 (P < 0.05 for 60 and 90 mg BID and 60 mg QD). Response (50% reduction) relative to placebo was achieved by 81.4% to 94.7% versus 58.5% of participants at end of treatment (all doses P < 0.05). Treatment-emergent adverse events were largely mild/moderate; no serious treatment-related treatment-emergent adverse events occurred. Conclusions: Fezolinetant is a well-tolerated, effective nonhormone therapy that rapidly reduces moderate/severe menopausal VMS. Video Summary:http://links.lww.com/MENO/A572; video script available at http://links.lww.com/MENO/A573.
Collapse
|
26
|
Delgado GA, Glazer RA, Brown-Peterson NJ. Arrested Sexual Development in Queen Conch ( Lobatus gigas) Linked to Abnormalities in the Cerebral Ganglia. THE BIOLOGICAL BULLETIN 2019; 237:241-249. [PMID: 31922908 DOI: 10.1086/706494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
In the Florida Keys, queen conchs (Lobatus gigas) occur in two spatially distinct regions: nearshore in habitats immediately adjacent to the shoreline and offshore in habitats along the reef tract south of the islands. Our previous research demonstrated that adult conchs nearshore are not reproductively active, showing deficiencies in their gonadal condition compared to their offshore counterparts. Because sexual development in gastropods is controlled by hormones secreted by the cerebral ganglia, we hypothesized that the reproductive deficiencies seen in nearshore queen conchs involved the cerebral ganglia. We collected nearshore and offshore adults and made histological comparisons of their gonads and cerebral ganglia. Our results confirmed that gonadal maturity was delayed and that gamete production was reduced in nearshore conchs compared to offshore animals. These gonadal deficiencies in nearshore conchs were associated with abnormal cerebral ganglion histology (i.e., significant hypertrophy of ganglion cells and significantly lower density of ganglion cells). In addition, the shells of nearshore conchs were significantly lighter, which is particularly consequential because shell formation in gastropods is also mediated by hormones secreted by the cerebral ganglia. Given these results, it is apparent that some yet unidentified factor(s) is interfering with hormone production in the cerebral ganglia, to the detriment of gonad development and shell formation.
Collapse
|
27
|
Matsuda F, Ohkura S, Magata F, Munetomo A, Chen J, Sato M, Inoue N, Uenoyama Y, Tsukamura H. Role of kisspeptin neurons as a GnRH surge generator: Comparative aspects in rodents and non-rodent mammals. J Obstet Gynaecol Res 2019; 45:2318-2329. [PMID: 31608564 DOI: 10.1111/jog.14124] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 08/30/2019] [Indexed: 02/01/2023]
Abstract
Ovulation is an essential phenomenon for reproduction in mammalian females along with follicular growth. It is well established that gonadal function is controlled by the neuroendocrine system called the hypothalamus-pituitary-gonadal (HPG) axis. Gonadotropin-releasing hormone (GnRH) neurons, localized in the hypothalamus, had been considered to be the head in governing the HPG axis for a long time until the discovery of kisspeptin. In females, induction of ovulation and folliculogenesis has been linked to a surge mode and pulse mode of GnRH releases, respectively. The mechanisms of how the two modes of GnRH are differently regulated had long remained elusive. The discovery of kisspeptin neurons, distributed in two hypothalamic nuclei, such as the arcuate nucleus in the caudal hypothalamus and preoptic area or the anteroventral periventricular nucleus in the rostral hypothalamic regions, and analyses of the detailed functions of kisspeptin neurons have led marked progress on the understanding of different mechanisms regulating GnRH surges (ovulation) and GnRH pulses (folliculogenesis). The present review will focus on the role of kisspeptin neurons as the GnRH surge generator, including the sexual differentiation of the surge generation system and factors that regulate the surge generator. Comparative aspects between mammalian species are especially focused on.
Collapse
Affiliation(s)
- Fuko Matsuda
- Laboratory of Theriogenology, Department of Veterinary Medical Sciences, The University of Tokyo, Tokyo, Japan
| | - Satoshi Ohkura
- Laboratory of Animal Production Science, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Fumie Magata
- Laboratory of Theriogenology, Department of Veterinary Medical Sciences, The University of Tokyo, Tokyo, Japan
| | - Arisa Munetomo
- Laboratory of Theriogenology, Department of Veterinary Medical Sciences, The University of Tokyo, Tokyo, Japan
| | - Jing Chen
- Laboratory of Theriogenology, Department of Veterinary Medical Sciences, The University of Tokyo, Tokyo, Japan
| | - Marimo Sato
- Laboratory of Theriogenology, Department of Veterinary Medical Sciences, The University of Tokyo, Tokyo, Japan
| | - Naoko Inoue
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Yoshihisa Uenoyama
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Hiroko Tsukamura
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| |
Collapse
|
28
|
Krajewski-Hall SJ, Miranda Dos Santos F, McMullen NT, Blackmore EM, Rance NE. Glutamatergic Neurokinin 3 Receptor Neurons in the Median Preoptic Nucleus Modulate Heat-Defense Pathways in Female Mice. Endocrinology 2019; 160:803-816. [PMID: 30753503 PMCID: PMC6424091 DOI: 10.1210/en.2018-00934] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 02/04/2019] [Indexed: 01/06/2023]
Abstract
We have proposed that arcuate neurons coexpressing kisspeptin, neurokinin B, and dynorphin (KNDy neurons) contribute to hot flushes via projections to neurokinin 3 receptor (NK3R)-expressing neurons in the median preoptic nucleus (MnPO). To characterize the thermoregulatory role of MnPO NK3R neurons in female mice, we ablated these neurons using injections of saporin toxin conjugated to a selective NK3R agonist. Loss of MnPO NK3R neurons increased the core temperature (TCORE) during the light phase, with the frequency distributions indicating a regulated shift in the balance point. The increase in TCORE in the ablated mice occurred despite changes in the ambient temperature and regardless of estrogen status. We next determined whether an acute increase in ambient temperature or higher TCORE would induce Fos in preoptic enhanced green fluorescent protein (EGFP)-immunoreactive neurons in Tacr3-EGFP mice. Fos activation was increased in the MnPO but no induction of Fos was found in NK3R (EGFP-immunoreactive) neurons. Thus, MnPO NK3R neurons are not activated by warm thermosensors in the skin or viscera and are not warm-sensitive neurons. Finally, RNAscope was used to determine whether Tacr3 (NK3R) mRNA was coexpressed with vesicular glutamate transporter 2 or vesicular γ-aminobutyric acid (GABA) transporter mRNA, markers of glutamatergic and GABAergic neurotransmission, respectively. In the MnPO, 94% of NK3R neurons were glutamatergic, but in the adjacent medial preoptic area, 97% of NK3R neurons were GABAergic. Thus, NK3R neurons in the MnPO are glutamatergic and play a role in reducing TCORE but are not activated by warm thermal stimuli (internal or external). These findings suggest that KNDy neurons modulate thermosensory pathways for heat defense indirectly via a subpopulation of glutamatergic MnPO neurons that express NK3R.
Collapse
Affiliation(s)
| | | | - Nathaniel T McMullen
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, Arizona
| | - Elise M Blackmore
- Department of Pathology, University of Arizona College of Medicine, Tucson, Arizona
| | - Naomi E Rance
- Department of Pathology, University of Arizona College of Medicine, Tucson, Arizona
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, Arizona
- Department of Neurology, University of Arizona College of Medicine, Tucson, Arizona
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, Arizona
| |
Collapse
|
29
|
Hrabovszky E, Takács S, Göcz B, Skrapits K. New Perspectives for Anatomical and Molecular Studies of Kisspeptin Neurons in the Aging Human Brain. Neuroendocrinology 2019; 109:230-241. [PMID: 30612127 DOI: 10.1159/000496566] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/04/2019] [Indexed: 11/19/2022]
Abstract
The human infundibular nucleus (corresponding to the rodent arcuate nucleus) serves as an important integration center for neuronal signals and hormones released by peripheral endocrine organs. Kisspeptin (KP)-producing neurons of this anatomical site, many of which also synthesize neurokinin B (NKB), are critically involved in sex hormone signaling to gonadotropin-releasing hormone (GnRH) neurons. In recent years, the basic topography, morphology, neuropeptide content, and connectivity of human KP neurons have been investigated with in situ hybridization and immunohistochemistry on postmortem tissues. These studies revealed that human KP neurons differ neurochemically from their rodent counterparts and show robust aging-related plasticity. Earlier immunohistochemical experiments also provided evidence for temporal changes in the hypothalamus of aging men whose NKB and KP neurons undergo hypertrophy, increase in number, exhibit increased neuropeptide mRNA expression and immunoreactivity and give rise to higher numbers of immunoreactive fibers and afferent contacts onto GnRH neurons. Increasing percentages of KP-expressing NKB perikarya, NKB axons, and NKB inputs to GnRH neurons raise the intriguing possibility that a significant subset of NKB neurons begins to cosynthesize KP as aging advances. Although use of postmortem tissues is technically challenging, recently available single-cell anatomical and molecular approaches discussed in this review provide promising new tools to investigate the aging-related anatomical and functional plasticity of the human KP neuronal system.
Collapse
Affiliation(s)
- Erik Hrabovszky
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary,
| | - Szabolcs Takács
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Balázs Göcz
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Katalin Skrapits
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| |
Collapse
|
30
|
Szeliga A, Czyzyk A, Podfigurna A, Genazzani AR, Genazzani AD, Meczekalski B. The role of kisspeptin/neurokinin B/dynorphin neurons in pathomechanism of vasomotor symptoms in postmenopausal women: from physiology to potential therapeutic applications. Gynecol Endocrinol 2018; 34:913-919. [PMID: 29902942 DOI: 10.1080/09513590.2018.1480711] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Women during perimenopausal period experience a range of symptoms, which interfere with physical, sexual, and social life. About 65-75% of symptoms connected with postmenopausal period are vasomotor symptoms (VMS), such as hot flushes and night sweats. Hot flushes are subjective sensation of heat associated with cutaneous vasodilatation and drop in core temperature. It is suspected that VMS are strongly correlated with pulsatile oversecretion of gonadotropin-releasing hormone (GnRH) and subsequently luteinizing hormone (LH). Evidence has accumulated in parallel showing that lack of negative feedback of steroid hormones synthesized in ovary causes overactivation of hypertrophied kisspeptin/neurokinin B/dynorphin (KNDy) neurons, located in infundibular nucleus. Oversecretion of both kisspeptin (KISS1) and neurokinin B (NKB), as well as downregulation of dynorphin, plays dominant role in creation of GnRH pulses. This in turn causes VMS. Administration of senktide, highly potent and selective NK3R agonist, resulted in increase of serum LH concentration, induction of VMS, increase in heart rate, and skin temperature in postmenopausal women. These finding suggest that modulation of KNDy neurons may become new therapeutic approach in the treatment of VMS.
Collapse
Affiliation(s)
- Anna Szeliga
- a Department of Gynecological Endocrinology , Poznan University of Medical Sciences , Poznan , Poland
| | - Adam Czyzyk
- a Department of Gynecological Endocrinology , Poznan University of Medical Sciences , Poznan , Poland
| | - Agnieszka Podfigurna
- a Department of Gynecological Endocrinology , Poznan University of Medical Sciences , Poznan , Poland
| | - Andrea R Genazzani
- b Department of Reproductive Medicine and Child Development, Division of Gynecology and Obstetrics , University of Pisa , Pisa , Italy
| | - Alessandro D Genazzani
- c Department of Obstetrics and Gynecology , Gynecological Endocrinology Center, University of Modena and Reggio Emilia , Modena , Italy
| | - Blazej Meczekalski
- a Department of Gynecological Endocrinology , Poznan University of Medical Sciences , Poznan , Poland
| |
Collapse
|
31
|
Miragem AA, Homem de Bittencourt PI. Nitric oxide-heat shock protein axis in menopausal hot flushes: neglected metabolic issues of chronic inflammatory diseases associated with deranged heat shock response. Hum Reprod Update 2018; 23:600-628. [PMID: 28903474 DOI: 10.1093/humupd/dmx020] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 06/28/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Although some unequivocal underlying mechanisms of menopausal hot flushes have been demonstrated in animal models, the paucity of similar approaches in humans impedes further mechanistic outcomes. Human studies might show some as yet unexpected physiological mechanisms of metabolic adaptation that permeate the phase of decreased oestrogen levels in both symptomatic and asymptomatic women. This is particularly relevant because both the severity and time span of hot flushes are associated with increased risk of chronic inflammatory disease. On the other hand, oestrogen induces the expression of heat shock proteins of the 70 kDa family (HSP70), which are anti-inflammatory and cytoprotective protein chaperones, whose expression is modulated by different types of physiologically stressful situations, including heat stress and exercise. Therefore, lower HSP70 expression secondary to oestrogen deficiency increases cardiovascular risk and predisposes the patient to senescence-associated secretory phenotype (SASP) that culminates in chronic inflammatory diseases, such as obesities, type 2 diabetes, neuromuscular and neurodegenerative diseases. OBJECTIVE AND RATIONALE This review focuses on HSP70 and its accompanying heat shock response (HSR), which is an anti-inflammatory and antisenescent pathway whose intracellular triggering is also oestrogen-dependent via nitric oxide (NO) production. The main goal of the manuscript was to show that the vasomotor symptoms that accompany hot flushes may be a disguised clue for important neuroendocrine alterations linking oestrogen deficiency to the anti-inflammatory HSR. SEARCH METHODS Results from our own group and recent evidence on hypothalamic control of central temperature guided a search on PubMed and Google Scholar websites. OUTCOMES Oestrogen elicits rapid production of the vasodilatory gas NO, a powerful activator of HSP70 expression. Whence, part of the protective effects of oestrogen over cardiovascular and neuroendocrine systems is tied to its capacity of inducing the NO-elicited HSR. The hypothalamic areas involved in thermoregulation (infundibular nucleus in humans and arcuate nucleus in other mammals) and whose neurons are known to have their function altered after long-term oestrogen ablation, particularly kisspeptin-neurokinin B-dynorphin neurons, (KNDy) are the same that drive neuroprotective expression of HSP70 and, in many cases, this response is via NO even in the absence of oestrogen. From thence, it is not illogical that hot flushes might be related to an evolutionary adaptation to re-equip the NO-HSP70 axis during the downfall of circulating oestrogen. WIDER IMPLICATIONS Understanding of HSR could shed light on yet uncovered mechanisms of menopause-associated diseases as well as on possible manipulation of HSR in menopausal women through physiological, pharmacological, nutraceutical and prebiotic interventions. Moreover, decreased HSR indices (that can be clinically determined with ease) in perimenopause could be of prognostic value in predicting the moment and appropriateness of starting a HRT.
Collapse
Affiliation(s)
- Antônio Azambuja Miragem
- Laboratory of Cellular Physiology, Department of Physiology, Federal University of Rio Grande do Sul, Rua Sarmento Leite 500, ICBS, 2nd Floor, Suite 350, Porto Alegre, RS 90050-170, Brazil.,Federal Institute of Education, Science and Technology 'Farroupilha', Rua Uruguai 1675, Santa Rosa, RS 98900-000, Brazil
| | - Paulo Ivo Homem de Bittencourt
- Laboratory of Cellular Physiology, Department of Physiology, Federal University of Rio Grande do Sul, Rua Sarmento Leite 500, ICBS, 2nd Floor, Suite 350, Porto Alegre, RS 90050-170, Brazil
| |
Collapse
|
32
|
Prague JK, Dhillo WS. Neurokinin 3 receptor antagonism – the magic bullet for hot flushes? Climacteric 2017; 20:505-509. [DOI: 10.1080/13697137.2017.1385598] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- J. K. Prague
- Department of Investigative Medicine, Imperial College London, London, UK
| | - W. S. Dhillo
- Department of Investigative Medicine, Imperial College London, London, UK
| |
Collapse
|
33
|
Contrôle de l’axe gonadotrope : nouveaux aspects physiologiques et thérapeutiques. ANNALES D'ENDOCRINOLOGIE 2017; 78 Suppl 1:S31-S40. [DOI: 10.1016/s0003-4266(17)30923-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
34
|
Sturdee DW, Hunter MS, Maki PM, Gupta P, Sassarini J, Stevenson JC, Lumsden MA. The menopausal hot flush: a review. Climacteric 2017; 20:296-305. [DOI: 10.1080/13697137.2017.1306507] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- D. W. Sturdee
- Department of Gynaecology, Solihull Hospital, Birmingham, UK
| | - M. S. Hunter
- Unit of Psychology, Institute of Psychiatry (at Guy's), King's College London, London, UK
| | - P. M. Maki
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| | - P. Gupta
- Women's Unit, Solihull Hospital, Birmingham Heartlands & Solihull NHS Trust, Birmingham, UK
| | - J. Sassarini
- Department of Obstetrics & Gynaecology, University of Glasgow, College of Medicine, Western Infirmary, Glasgow, UK
| | - J. C. Stevenson
- National Heart & Lung Institute, Royal Brompton Hospital, London, UK
| | - M. A. Lumsden
- Reproductive and Maternal Medicine, University of Glasgow, Glasgow, UK
| |
Collapse
|
35
|
Depression in midlife women. Maturitas 2016; 94:149-154. [DOI: 10.1016/j.maturitas.2016.09.004] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 08/30/2016] [Accepted: 09/04/2016] [Indexed: 12/26/2022]
|
36
|
Mittelman-Smith MA, Krajewski-Hall SJ, McMullen NT, Rance NE. Ablation of KNDy Neurons Results in Hypogonadotropic Hypogonadism and Amplifies the Steroid-Induced LH Surge in Female Rats. Endocrinology 2016; 157:2015-27. [PMID: 26937713 PMCID: PMC4870865 DOI: 10.1210/en.2015-1740] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In the human infundibular (arcuate) nucleus, a subpopulation of neurons coexpress kisspeptin and neurokinin B (NKB), 2 peptides required for normal reproductive function. A homologous group of neurons exists in the arcuate nucleus of rodents, termed KNDy neurons based on the coexpression of kisspeptin, NKB, and dynorphin. To study their function, we recently developed a method to selectively ablate KNDy neurons using NK3-SAP, a neurokinin 3 receptor agonist conjugated to saporin (SAP). Here, we ablated KNDy neurons in female rats to determine whether these neurons are required for estrous cyclicity and the steroid induced LH surge. NK3-SAP or Blank-SAP (control) was microinjected into the arcuate nucleus using stereotaxic surgery. After monitoring vaginal smears for 3-4 weeks, rats were ovariectomized and given 17β-estradiol and progesterone in a regimen that induced an afternoon LH surge. Rats were killed at the time of peak LH levels, and brains were harvested for NKB and dual labeled GnRH/Fos immunohistochemistry. In ovary-intact rats, ablation of KNDy neurons resulted in hypogonadotropic hypogonadism, characterized by low levels of serum LH, constant diestrus, ovarian atrophy with increased follicular atresia, and uterine atrophy. Surprisingly, the 17β-estradiol and progesterone-induced LH surge was 3 times higher in KNDy-ablated rats. Despite the marked increase in the magnitude of the LH surge, the number of GnRH or anterior ventral periventricular nucleus neurons expressing Fos was not significantly different between groups. Our studies show that KNDy neurons are essential for tonic levels of serum LH and estrous cyclicity and may play a role in limiting the magnitude of the LH surge.
Collapse
Affiliation(s)
- Melinda A Mittelman-Smith
- Departments of Pathology (M.A.M.-S., S.J.K.-H., N.E.R.) and Cellular and Molecular Medicine and Neurology (N.T.M., N.E.R.) and The Evelyn F. McKnight Brain Institute (N.E.R.), University of Arizona College of Medicine, Tucson, Arizona 85724
| | - Sally J Krajewski-Hall
- Departments of Pathology (M.A.M.-S., S.J.K.-H., N.E.R.) and Cellular and Molecular Medicine and Neurology (N.T.M., N.E.R.) and The Evelyn F. McKnight Brain Institute (N.E.R.), University of Arizona College of Medicine, Tucson, Arizona 85724
| | - Nathaniel T McMullen
- Departments of Pathology (M.A.M.-S., S.J.K.-H., N.E.R.) and Cellular and Molecular Medicine and Neurology (N.T.M., N.E.R.) and The Evelyn F. McKnight Brain Institute (N.E.R.), University of Arizona College of Medicine, Tucson, Arizona 85724
| | - Naomi E Rance
- Departments of Pathology (M.A.M.-S., S.J.K.-H., N.E.R.) and Cellular and Molecular Medicine and Neurology (N.T.M., N.E.R.) and The Evelyn F. McKnight Brain Institute (N.E.R.), University of Arizona College of Medicine, Tucson, Arizona 85724
| |
Collapse
|
37
|
Cernea M, Padmanabhan V, Goodman RL, Coolen LM, Lehman MN. Prenatal Testosterone Treatment Leads to Changes in the Morphology of KNDy Neurons, Their Inputs, and Projections to GnRH Cells in Female Sheep. Endocrinology 2015; 156:3277-91. [PMID: 26061725 PMCID: PMC4541615 DOI: 10.1210/en.2014-1609] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Prenatal testosterone (T)-treated ewes display a constellation of reproductive defects that closely mirror those seen in PCOS women, including altered hormonal feedback control of GnRH. Kisspeptin/neurokinin B/dynorphin (KNDy) neurons of the arcuate nucleus (ARC) play a key role in steroid feedback control of GnRH secretion, and prenatal T treatment in sheep causes an imbalance of KNDy peptide expression within the ARC. In the present study, we tested the hypothesis that prenatal T exposure, in addition to altering KNDy peptides, leads to changes in the morphology and synaptic inputs of this population, kisspeptin cells of the preoptic area (POA), and GnRH cells. Prenatal T treatment significantly increased the size of KNDy cell somas, whereas POA kisspeptin, GnRH, agouti-related peptide, and proopiomelanocortin neurons were each unchanged in size. Prenatal T treatment also significantly reduced the total number of synaptic inputs onto KNDy neurons and POA kisspeptin neurons; for KNDy neurons, the decrease was partly due to a decrease in KNDy-KNDy synapses, whereas KNDy inputs to POA kisspeptin cells were unaltered. Finally, prenatal T reduced the total number of inputs to GnRH cells in both the POA and medial basal hypothalamus, and this change was in part due to a decreased number of inputs from KNDy neurons. The hypertrophy of KNDy cells in prenatal T sheep resembles that seen in ARC kisspeptin cells of postmenopausal women, and together with changes in their synaptic inputs and projections to GnRH neurons, may contribute to defects in steroidal control of GnRH observed in this animal model.
Collapse
Affiliation(s)
- Maria Cernea
- Department of Neurobiology and Anatomical Sciences (M.C., L.M.C., M.N.L.), The University of Mississippi Medical Center, Jackson, Mississippi 39232; Department of Obstetrics and Gynecology, Pediatrics, and Reproductive Sciences Program (V.P.), University of Michigan, Ann Arbor, Michigan 48109; and Department of Physiology and Pharmacology (R.L.G.), West Virginia University, Morgantown, West Virginia 26506
| | - Vasantha Padmanabhan
- Department of Neurobiology and Anatomical Sciences (M.C., L.M.C., M.N.L.), The University of Mississippi Medical Center, Jackson, Mississippi 39232; Department of Obstetrics and Gynecology, Pediatrics, and Reproductive Sciences Program (V.P.), University of Michigan, Ann Arbor, Michigan 48109; and Department of Physiology and Pharmacology (R.L.G.), West Virginia University, Morgantown, West Virginia 26506
| | - Robert L Goodman
- Department of Neurobiology and Anatomical Sciences (M.C., L.M.C., M.N.L.), The University of Mississippi Medical Center, Jackson, Mississippi 39232; Department of Obstetrics and Gynecology, Pediatrics, and Reproductive Sciences Program (V.P.), University of Michigan, Ann Arbor, Michigan 48109; and Department of Physiology and Pharmacology (R.L.G.), West Virginia University, Morgantown, West Virginia 26506
| | - Lique M Coolen
- Department of Neurobiology and Anatomical Sciences (M.C., L.M.C., M.N.L.), The University of Mississippi Medical Center, Jackson, Mississippi 39232; Department of Obstetrics and Gynecology, Pediatrics, and Reproductive Sciences Program (V.P.), University of Michigan, Ann Arbor, Michigan 48109; and Department of Physiology and Pharmacology (R.L.G.), West Virginia University, Morgantown, West Virginia 26506
| | - Michael N Lehman
- Department of Neurobiology and Anatomical Sciences (M.C., L.M.C., M.N.L.), The University of Mississippi Medical Center, Jackson, Mississippi 39232; Department of Obstetrics and Gynecology, Pediatrics, and Reproductive Sciences Program (V.P.), University of Michigan, Ann Arbor, Michigan 48109; and Department of Physiology and Pharmacology (R.L.G.), West Virginia University, Morgantown, West Virginia 26506
| |
Collapse
|
38
|
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
|
39
|
Mittelman-Smith MA, Krajewski-Hall SJ, McMullen NT, Rance NE. Neurokinin 3 Receptor-Expressing Neurons in the Median Preoptic Nucleus Modulate Heat-Dissipation Effectors in the Female Rat. Endocrinology 2015; 156:2552-62. [PMID: 25825817 PMCID: PMC4475724 DOI: 10.1210/en.2014-1974] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
KNDy neurons facilitate tail skin vasodilation and modulate the effects of estradiol on thermoregulation. We hypothesize that KNDy neurons influence cutaneous vasodilation via projections to neurons in the median preoptic nucleus (MnPO) that express the neurokinin 3 receptor (NK3R). In support of this hypothesis, focal microinjections of senktide, an NK3R agonist, into the MnPO lowers core temperature (TCORE) in the female rat. To further study the role of MnPO NK3R neurons in thermoregulation, these neurons were specifically ablated using a conjugate of a selective NK3R agonist and saporin (NK3-SAP). NK3-SAP or blank-SAP (control) was injected into the MnPO/medial septum. Tail skin temperature (TSKIN) and TCORE were measured in ovariectomized rats exposed to 3 ambient temperatures (TAMBIENT) before and after estradiol-17β (E2) treatment. Before killing, we injected senktide (sc), monitored TCORE for 70 minutes, and harvested brains for Fos immunohistochemistry. Ablation of MnPO NK3R neurons lowered TSKIN at neutral and subneutral TAMBIENT regardless of E2 treatment. However, ablation did not prevent the effects of E2 on TCORE and TSKIN. In control rats, senktide injections induced hypothermia with numerous Fos-immunoreactive cells in the MnPO. In contrast, in NK3-SAP rats, senktide did not alter TCORE and minimal Fos-immunoreactive neurons were identified in the MnPO. These data show that NK3R neurons in the MnPO are required for the hypothermic effects of senktide but not for the E2 modulation of thermoregulation. The lower TSKIN in NK3-SAP-injected rats suggests that MnPO NK3R neurons, like KNDy neurons, facilitate cutaneous vasodilation, an important heat-dissipation effector.
Collapse
Affiliation(s)
- Melinda A Mittelman-Smith
- Departments of Pathology (M.A.M.-S., S.J.K.-H., N.E.R.), Cellular and Molecular Medicine (N.T.M., N.E.R.), and Neurology (N.E.R.), and the Evelyn F. McKnight Brain Institute (N.E.R.), University of Arizona College of Medicine, Tucson, Arizona 85724
| | - Sally J Krajewski-Hall
- Departments of Pathology (M.A.M.-S., S.J.K.-H., N.E.R.), Cellular and Molecular Medicine (N.T.M., N.E.R.), and Neurology (N.E.R.), and the Evelyn F. McKnight Brain Institute (N.E.R.), University of Arizona College of Medicine, Tucson, Arizona 85724
| | - Nathaniel T McMullen
- Departments of Pathology (M.A.M.-S., S.J.K.-H., N.E.R.), Cellular and Molecular Medicine (N.T.M., N.E.R.), and Neurology (N.E.R.), and the Evelyn F. McKnight Brain Institute (N.E.R.), University of Arizona College of Medicine, Tucson, Arizona 85724
| | - Naomi E Rance
- Departments of Pathology (M.A.M.-S., S.J.K.-H., N.E.R.), Cellular and Molecular Medicine (N.T.M., N.E.R.), and Neurology (N.E.R.), and the Evelyn F. McKnight Brain Institute (N.E.R.), University of Arizona College of Medicine, Tucson, Arizona 85724
| |
Collapse
|
40
|
Gawałek M, Sliwowska JH. Neuronal basis of reproductive dysfunctions associated with diet and alcohol: From the womb to adulthood. Reprod Biol 2015; 15:69-78. [PMID: 26051454 DOI: 10.1016/j.repbio.2015.04.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Revised: 04/08/2015] [Accepted: 04/14/2015] [Indexed: 12/19/2022]
Abstract
The theory that individuals are born as tabula rasa and that their knowledge comes from experience and perception is no longer true. Studies suggest that experience is gained as early as in the mother's womb. Moreover, environmental stressors like alcohol or inadequate diet can affect physiological systems such as the hypothalmic-pituitary-gonadal (HPG) axis. The effects of these stressors can manifest as alterations in sexual development and adult reproductive functions. In this review, we consider and compare evidence from animal models and human studies demonstrating the role of environmental stressors (alcohol and under- or overnutrition) on the HPG axis. We review the role of alcohol and inadequate diet in prenatal reproductive system programming and consider specific candidate neurons in the adult hypothalamus through which reproductive function is being regulated. Finally, we review evidence from animal studies on the role that alcohol and diet play in fertility and reproductive disorders. We conclude that in order to better understand reproductive failure in animals and humans we need to consider in utero development and pay more attention to early life experience when searching for the origins of reproductive diseases.
Collapse
Affiliation(s)
- Monika Gawałek
- Laboratory of Neurobiology, Institute of Zoology, Poznań University of Life Sciences, Wojska Polskiego 71C, 60-625 Poznań, Poland.
| | - Joanna H Sliwowska
- Laboratory of Neurobiology, Institute of Zoology, Poznań University of Life Sciences, Wojska Polskiego 71C, 60-625 Poznań, Poland.
| |
Collapse
|
41
|
Cholanian M, Krajewski-Hall SJ, McMullen NT, Rance NE. Chronic oestradiol reduces the dendritic spine density of KNDy (kisspeptin/neurokinin B/dynorphin) neurones in the arcuate nucleus of ovariectomised Tac2-enhanced green fluorescent protein transgenic mice. J Neuroendocrinol 2015; 27:253-63. [PMID: 25659412 PMCID: PMC4788980 DOI: 10.1111/jne.12263] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 01/08/2015] [Accepted: 02/03/2015] [Indexed: 11/27/2022]
Abstract
Neurones in the arcuate nucleus that express neurokinin B (NKB), kisspeptin and dynorphin (KNDy) play an important role in the reproductive axis. Oestradiol modulates the gene expression and somatic size of these neurones, although there is limited information available about whether their dendritic structure, a correlate of cellular plasticity, is altered by oestrogens. In the present study, we investigated the morphology of KNDy neurones by filling fluorescent neurones in the arcuate nucleus of Tac2-enhanced green fluorescent protein (EGFP) transgenic mice with biocytin. Filled neurones from ovariectomised (OVX) or OVX plus 17β-oestradiol (E2)-treated mice were visualised with anti-biotin immunohistochemistry and reconstructed in three dimensions with computer-assisted microscopy. KNDy neurones exhibited two primary dendrites, each with a few branches confined to the arcuate nucleus. Quantitative analysis revealed that E2 treatment of OVX mice decreased the cell size and dendritic spine density of KNDy neurones. The axons of KNDy neurones originated from the cell body or proximal dendrite and gave rise to local branches that appeared to terminate within the arcuate nucleus. Numerous terminal boutons were also visualised within the ependymal layer of the third ventricle adjacent to the arcuate nucleus. Axonal branches also projected to the adjacent median eminence and exited the arcuate nucleus. Confocal microscopy revealed close apposition of EGFP and gonadotrophin-releasing hormone-immunoreactive fibres within the median eminence and confirmed the presence of KNDy axon terminals in the ependymal layer of the third ventricle. The axonal branching pattern of KNDy neurones suggests that a single KNDy neurone could influence multiple arcuate neurones, tanycytes in the wall of the third ventricle, axon terminals in the median eminence and numerous areas outside of the arcuate nucleus. In parallel with its inhibitory effects on electrical excitability, E2 treatment of OVX Tac2-EGFP mice induces structural changes in the somata and dendrites of KNDy neurones.
Collapse
Affiliation(s)
- Marina Cholanian
- Department of Pathology, University of Arizona College of Medicine, Tucson, Arizona, USA
| | | | - Nathaniel T. McMullen
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Naomi E. Rance
- Department of Pathology, Neurology and the Evelyn F. McKnight Brain Institute University of Arizona College of Medicine, Tucson, AZ, USA
- CORRESPONDENCE TO: Naomi E. Rance, MD, PhD, Department of Pathology, University of Arizona College of Medicine, 1501 N. Campbell Ave, Tucson, AZ 85724, USA, , phone: (520) 626-6099
| |
Collapse
|
42
|
True C, Nasrin Alam S, Cox K, Chan YM, Seminara SB. Neurokinin B is critical for normal timing of sexual maturation but dispensable for adult reproductive function in female mice. Endocrinology 2015; 156:1386-97. [PMID: 25574869 PMCID: PMC4399316 DOI: 10.1210/en.2014-1862] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Humans carrying mutations in neurokinin B (NKB) or the NKB receptor fail to undergo puberty due to decreased secretion of GnRH. Despite this pubertal delay, many of these patients go on to achieve activation of their hypothalamic-pituitary-gonadal axis in adulthood, a phenomenon termed reversal, indicating that NKB signaling may play a more critical role for the timing of pubertal development than adult reproductive function. NKB receptor-deficient mice are hypogonadotropic but have no defects in the timing of sexual maturation. The current study has performed the first phenotypic evaluation of mice bearing mutations in Tac2, the gene encoding the NKB ligand, to determine whether they have impaired sexual development similar to their human counterparts. Male Tac2-/- mice showed no difference in the timing of sexual maturation or fertility compared with wild-type littermates and were fertile. In contrast, Tac2-/- females had profound delays in sexual maturation, with time to vaginal opening and first estrus occurring significantly later than controls, and initial abnormalities in estrous cycles. However, cycling recovered in adulthood and Tac2-/- females were fertile, although they produced fewer pups per litter. Thus, female Tac2-/- mice parallel humans harboring NKB pathway mutations, with delayed sexual maturation and activation of the reproductive cascade later in life. Moreover, direct comparison of NKB ligand and receptor-deficient females confirmed that only NKB ligand-deficient animals have delayed sexual maturation, suggesting that in the absence of the NKB receptor, NKB may regulate the timing of sexual maturation through other tachykinin receptors.
Collapse
Affiliation(s)
- Cadence True
- Harvard Reproductive Sciences Center and Reproductive Endocrine Unit (C.T., S.N.A., K.C., Y.-M.C., S.S.), Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114; and Division of Endocrinology (Y.-M.C.), Department of Medicine, Boston Children's Hospital, Boston, Massachusetts 02115
| | | | | | | | | |
Collapse
|
43
|
Eghlidi DH, Urbanski HF. Effects of Age and Estradiol on Gene Expression in the Rhesus Macaque Hypothalamus. Neuroendocrinology 2015; 101:236-45. [PMID: 25765287 PMCID: PMC4475460 DOI: 10.1159/000381063] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 02/18/2015] [Indexed: 11/19/2022]
Abstract
BACKGROUND The hypothalamus plays a key role in mediating the effects of estrogen on many physiological functions, including reproduction, metabolism, and thermoregulation. We have previously observed marked estrogen-dependent gene expression changes within the hypothalamus of rhesus macaques during aging, especially in the KNDy neurons of the arcuate-median eminence (ARC-ME) that produce kisspeptin, neurokinin B, and dynorphin A. Little is known, however, about the mechanisms involved in mediating the feedback from estrogen onto these neurons. METHODS We used quantitative real-time PCR to profile age- and estrogen-dependent gene expression changes in the rhesus macaque hypothalamus. Our focus was on genes that encode steroid receptors (ESR1, ESR2, PGR, and AR) and on enzymes that contribute to the local synthesis of 17β-estradiol (E2; STS, HSD3B1/2, HSD17B5, and CYP19A). In addition, we used RT(2) Profiler™ PCR Arrays to profile a larger set of genes that are integral to hypothalamic function. RESULTS KISS1, KISS1R, TAC3, and NPY2R mRNA levels increased in surgically menopausal (ovariectomized) old females relative to age-matched ovariectomized animals that received E2 hormone therapy. In contrast, PGR, HSD17B, GNRH2, SLC6A3, KISS1, TAC3, and NPY2R mRNA levels increased after E2 supplementation. CONCLUSION The rhesus macaque ARC-ME expresses many genes that are responsive to changes in circulating estrogen levels, even during old age, and these may contribute to causing the normal and pathophysiological changes that occur during menopause.
Collapse
Affiliation(s)
- Dominique H. Eghlidi
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oreg., USA
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, Oreg., USA
| | - Henryk F. Urbanski
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oreg., USA
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, Oreg., USA
- Deptartment of Physiology and Pharmacology, Oregon Health and Science University, Portland, Oreg., USA
| |
Collapse
|
44
|
Naugle MM, Nguyen LT, Merceron TK, Filardo E, Janssen WGM, Morrison JH, Rapp PR, Gore AC. G-protein coupled estrogen receptor, estrogen receptor α, and progesterone receptor immunohistochemistry in the hypothalamus of aging female rhesus macaques given long-term estradiol treatment. ACTA ACUST UNITED AC 2014; 321:399-414. [PMID: 24862737 DOI: 10.1002/jez.1871] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 04/28/2014] [Indexed: 12/15/2022]
Abstract
Steroid hormone receptors are widely and heterogeneously expressed in the brain, and are regulated by age and gonadal hormones. Our goal was to quantify effects of aging, long-term estradiol (E2 ) treatment, and their interactions, on expression of G protein-coupled estrogen receptor (GPER), estrogen receptor α (ERα) and progesterone receptor (PR) immunoreactivity in two hypothalamic regions, the arcuate (ARC) and the periventricular area (PERI) of rhesus monkeys as a model of menopause and hormone replacement. Ovariectomized (OVX) rhesus macaques were young (∼ 11 years) or aged (∼ 25 years), given oil (vehicle) or E2 every 3 weeks for 2 years. Immunohistochemistry and stereologic analysis of ERα, PR, and GPER was performed. More effects were detected for GPER than the other two receptors. Specifically, GPER cell density in the ARC and PERI, and the percent of GPER-immunoreactive cells in the PERI, were greater in aged than in young monkeys. In addition, we mapped the qualitative distribution of GPER in the monkey hypothalamus and nearby regions. For ERα, E2 treated monkeys tended to have higher cell density than vehicle monkeys in the ARC. The percent of PR density in the PERI tended to be higher in E2 than vehicle monkeys of both ages. This study shows that the aged hypothalamus maintains expression of hormone receptors with age, and that long-term cyclic E2 treatment has few effects on their expression, although GPER was affected more than ERα or PR. This result is surprising in light of evidence for E2 regulation of the receptors studied here, and differences may be due to the selected regions, long-term nature of E2 treatment, among other possibilities.
Collapse
Affiliation(s)
- Michelle M Naugle
- Institute for Neuroscience, University of Texas at Austin, Austin, Texas
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Skorupskaite K, George JT, Anderson RA. The kisspeptin-GnRH pathway in human reproductive health and disease. Hum Reprod Update 2014; 20:485-500. [PMID: 24615662 PMCID: PMC4063702 DOI: 10.1093/humupd/dmu009] [Citation(s) in RCA: 299] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The discovery of kisspeptin as key central regulator of GnRH secretion has led to a new level of understanding of the neuroendocrine regulation of human reproduction. The related discovery of the kisspeptin-neurokinin B-dynorphin (KNDy) pathway in the last decade has further strengthened our understanding of the modulation of GnRH secretion by endocrine, metabolic and environmental inputs. In this review, we summarize current understanding of the physiological roles of these novel neuropeptides, and discuss the clinical relevance of these discoveries and their potential translational applications. METHODS A systematic literature search was performed using PUBMED for all English language articles up to January 2014. In addition, the reference lists of all relevant original research articles and reviews were examined. This review focuses mainly on published human studies but also draws on relevant animal data. RESULTS Kisspeptin is a principal regulator of the secretion of gonadotrophins, and through this key role it is critical for the onset of puberty, the regulation of sex steroid-mediated feedback and the control of adult fertility. Although there is some sexual dimorphism, both neuroanatomically and functionally, these functions are apparent in both men and women. Kisspeptin acts upstream of GnRH and, following paracrine stimulatory and inhibitory inputs from neurokinin B and dynorphin (KNDy neuropeptides), signals directly to GnRH neurones to control pulsatile GnRH release. When administered to humans in different isoforms, routes and doses, kisspeptin robustly stimulates LH secretion and LH pulse frequency. Manipulation of the KNDy system is currently the focus of translational research with the possibility of future clinical application to regulate LH pulsatility, increasing gonadal sex steroid secretion in reproductive disorders characterized by decreased LH pulsatility, including hypothalamic amenorrhoea and hypogonadotropic hypogonadism. Conversely there may be scope to reduce the activity of the KNDy system to reduce LH secretion where hypersecretion of LH adds to the phenotype, such as in polycystic ovary syndrome. CONCLUSIONS Kisspeptin is a recently discovered neuromodulator that controls GnRH secretion mediating endocrine and metabolic inputs to the regulation of human reproduction. Manipulation of kisspeptin signalling has the potential for novel therapies in patients with pathologically low or high LH pulsatility.
Collapse
Affiliation(s)
- Karolina Skorupskaite
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Jyothis T George
- Diabetes Trials Unit, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Headington, Oxford OX3 7LJ, UK
| | - Richard A Anderson
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| |
Collapse
|
46
|
Hrabovszky E. Neuroanatomy of the human hypothalamic kisspeptin system. Neuroendocrinology 2014; 99:33-48. [PMID: 24401651 DOI: 10.1159/000356903] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 10/30/2013] [Indexed: 11/19/2022]
Abstract
Hypothalamic kisspeptin (KP) neurons are key players in the neuronal network that regulates the onset of puberty and the pulsatile secretion of gonadotropin-releasing hormone (GnRH). In various mammalian species, the majority of KP-synthesizing neurons are concentrated in two distinct cell populations in the preoptic region and the arcuate nucleus (ARC). While studies of female rodents have provided evidence that preoptic KP neurons play a critical sex-specific role in positive estrogen feedback, KP neurons of the ARC have been implicated in negative sex steroid feedback and they have also been hypothesized to contribute to the pulse generator network which regulates episodic GnRH secretion in both females and males. Except for relatively few morphological studies available in monkeys and humans, our neuroanatomical knowledge of the hypothalamic KP systems is predominantly based on observations of laboratory species which are phylogenetically distant from the human. This review article discusses the currently available literature on the topographic distribution, network connectivity, neurochemistry, sexual dimorphism, and aging-dependent morphological plasticity of the human hypothalamic KP neuronal system.
Collapse
Affiliation(s)
- Erik Hrabovszky
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| |
Collapse
|
47
|
Rance NE, Dacks PA, Mittelman-Smith MA, Romanovsky AA, Krajewski-Hall SJ. Modulation of body temperature and LH secretion by hypothalamic KNDy (kisspeptin, neurokinin B and dynorphin) neurons: a novel hypothesis on the mechanism of hot flushes. Front Neuroendocrinol 2013; 34:211-27. [PMID: 23872331 PMCID: PMC3833827 DOI: 10.1016/j.yfrne.2013.07.003] [Citation(s) in RCA: 230] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Revised: 07/09/2013] [Accepted: 07/12/2013] [Indexed: 12/31/2022]
Abstract
Despite affecting millions of individuals, the etiology of hot flushes remains unknown. Here we review the physiology of hot flushes, CNS pathways regulating heat-dissipation effectors, and effects of estrogen on thermoregulation in animal models. Based on the marked changes in hypothalamic kisspeptin, neurokinin B and dynorphin (KNDy) neurons in postmenopausal women, we hypothesize that KNDy neurons play a role in the mechanism of flushes. In the rat, KNDy neurons project to preoptic thermoregulatory areas that express the neurokinin 3 receptor (NK3R), the primary receptor for NKB. Furthermore, activation of NK₃R in the median preoptic nucleus, part of the heat-defense pathway, reduces body temperature. Finally, ablation of KNDy neurons reduces cutaneous vasodilatation and partially blocks the effects of estrogen on thermoregulation. These data suggest that arcuate KNDy neurons relay estrogen signals to preoptic structures regulating heat-dissipation effectors, supporting the hypothesis that KNDy neurons participate in the generation of flushes.
Collapse
Affiliation(s)
- Naomi E Rance
- Department of Pathology and the Evelyn F. McKnight Brain Research Institute, University of Arizona College of Medicine, Tucson, AZ 85724, USA.
| | | | | | | | | |
Collapse
|
48
|
Abstract
Decreased metabolic rate may precede cognitive impairment in Alzheimer's disease (AD) and is thus an early occurring hallmark. Several observations in post-mortem brain indicate that activated neurons are better able to withstand aging and AD, a phenomenon paraphrased by us as 'use it or lose it'. Moreover, a number of pharmacological and nonpharmacological studies support the concept that activation of the brain has beneficial effects and may to a certain degree restore several aspects of cognition and other central functions. For instance, the circadian system may be restimulated in Alzheimer patients by exposing them to more light or transcutaneous nerve stimulation. A procedure has been developed to culture human post-mortem brain tissue that allows testing of the efficacy of putative stimulatory compounds such as neurotrophins.
Collapse
Affiliation(s)
- Dick F Swaab
- Netherlands Institute for Brain Research, Meibergdreef 33, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|
49
|
Mittelman-Smith MA, Williams H, Krajewski-Hall SJ, Lai J, Ciofi P, McMullen NT, Rance NE. Arcuate kisspeptin/neurokinin B/dynorphin (KNDy) neurons mediate the estrogen suppression of gonadotropin secretion and body weight. Endocrinology 2012; 153:2800-12. [PMID: 22508514 PMCID: PMC3359616 DOI: 10.1210/en.2012-1045] [Citation(s) in RCA: 139] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 03/21/2012] [Indexed: 01/08/2023]
Abstract
Estrogen withdrawal increases gonadotropin secretion and body weight, but the critical cell populations mediating these effects are not well understood. Recent studies have focused on a subpopulation of hypothalamic arcuate neurons that coexpress estrogen receptor α, neurokinin 3 receptor (NK(3)R), kisspeptin, neurokinin B, and dynorphin for the regulation of reproduction. To investigate the function of kisspeptin/neurokinin B/dynorphin (KNDy) neurons, a novel method was developed to ablate these cells using a selective NK(3)R agonist conjugated to the ribosome-inactivating toxin, saporin (NK(3)-SAP). Stereotaxic injections of NK(3)-SAP in the arcuate nucleus ablated KNDy neurons, as demonstrated by the near-complete loss of NK(3)R, NKB, and kisspeptin-immunoreactive (ir) neurons and depletion of the majority of arcuate dynorphin-ir neurons. Selectivity was demonstrated by the preservation of proopiomelanocortin, neuropeptide Y, and GnRH-ir elements in the arcuate nucleus and median eminence. In control rats, ovariectomy (OVX) markedly increased serum LH, FSH, and body weight, and these parameters were subsequently decreased by treatment with 17β-estradiol. KNDy neuron ablation prevented the rise in serum LH after OVX and attenuated the rise in serum FSH. KNDy neuron ablation did not completely block the suppressive effects of E(2) on gonadotropin secretion, a finding consistent with redundant pathways for estrogen negative feedback. However, regardless of estrogen status, KNDy-ablated rats had lower levels of serum gonadotropins compared with controls. Surprisingly, KNDy neuron ablation prevented the dramatic effects of OVX and 17β-estradiol (E(2)) replacement on body weight and abdominal girth. These data provide evidence that arcuate KNDy neurons are essential for tonic gonadotropin secretion, the rise in LH after removal of E(2), and the E(2) modulation of body weight.
Collapse
Affiliation(s)
- Melinda A Mittelman-Smith
- Department of Pathology and the Evelyn F. McKnight Brain Institute, University of Arizona College of Medicine, Tucson, Arizona 85724, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
In the past two decades, an increasing body of evidence has demonstrated that several G protein-coupled receptor (GPCR)-ligand pairs are critical for normal human reproductive development and function. Patients harboring genetic insults in either the receptors or their cognate ligands have presented with reproductive disorders characterized by varying degrees of GnRH deficiency. These disorders include idiopathic hypogonadotropic hypogonadism (IHH) and Kallmann Syndrome (KS). Conversely, mutations in some of these ligand-receptor pairs have been associated with accelerated reproductive maturation, manifested as central precocious puberty (CPP). To date, a series of elegant studies have characterized four GPCRs that play important roles in the neuroendocrine control of human reproductive development and function: GnRHR, KISS1R, PROKR2 and NK3R. Furthermore, these studies provide insights into the mechanisms by which mutations in these receptors give rise to reproductive disease phenotypes. This report will review mutations identified in GPCRs involved in the neuroendocrine control of the human reproductive axis with the aims of elucidating structure-function relationships of these GPCRs and identifying correlations between these structure-function relationships and the genotypic-phenotypic characterization of the patients.
Collapse
Affiliation(s)
- Sekoni D Noel
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|