1
|
Faure MC, Corona R, Roomans C, Lenfant F, Foidart JM, Cornil CA. Role of Membrane Estrogen Receptor Alpha on the Positive Feedback of Estrogens on Kisspeptin and GnRH Neurons. eNeuro 2024; 11:ENEURO.0271-23.2024. [PMID: 39375032 PMCID: PMC11520851 DOI: 10.1523/eneuro.0271-23.2024] [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: 08/02/2023] [Revised: 06/14/2024] [Accepted: 09/23/2024] [Indexed: 10/09/2024] Open
Abstract
Estrogens act through nuclear and membrane-initiated signaling. Estrogen receptor alpha (ERα) is critical for reproduction, but the relative contribution of its nuclear and membrane signaling to the central regulation of reproduction is unclear. To address this question, two complementary approaches were used: estetrol (E4) a natural estrogen acting as an agonist of nuclear ERs, but as an antagonist of their membrane fraction, and the C451A-ERα mouse lacking mERα. E4 dose- dependently blocks ovulation in female rats, but the central mechanism underlying this effect is unknown. To determine whether E4 acts centrally to control ovulation, its effect was tested on the positive feedback of estradiol (E2) on neural circuits underlying luteinizing hormone (LH) secretion. In ovariectomized females chronically exposed to a low dose of E2, estradiol benzoate (EB) alone or combined with progesterone (P) induced an increase in the number of kisspeptin (Kp) and gonadotropin-releasing hormone (GnRH) neurons coexpressing Fos, a marker of neuronal activation. E4 blocked these effects of EB, but not when combined to P. These results indicate that E4 blocked the central induction of the positive feedback in the absence of P, suggesting an antagonistic effect of E4 on mERα in the brain as shown in peripheral tissues. In parallel, as opposed to wild-type females, C451A-ERα females did not show the activation of Kp and GnRH neurons in response to EB unless they are treated with P. Together these effects support a role for membrane-initiated estrogen signaling in the activation of the circuit mediating the LH surge.
Collapse
Affiliation(s)
- Mélanie C. Faure
- Laboratory of Neuroendocrinology, GIGA Neurosciences, University of Liège, Liège, Belgium
| | - Rebeca Corona
- Laboratory of Neuroendocrinology, GIGA Neurosciences, University of Liège, Liège, Belgium
| | - Céline Roomans
- Laboratory of Neuroendocrinology, GIGA Neurosciences, University of Liège, Liège, Belgium
| | - Françoise Lenfant
- Institute of Metabolic and Cardiovascular Diseases (I2MC) Equipe 4, Inserm U1297-UPS, CHU, Toulouse 31432, France
| | - Jean-Michel Foidart
- Department of Obstetrics and Gynecology, University of Liège, Liège, Belgium
- Estetra SRL, Légiapark, Boulevard Patience et Beaujonc 3, 4000 Liège, Belgium
| | - Charlotte A. Cornil
- Laboratory of Neuroendocrinology, GIGA Neurosciences, University of Liège, Liège, Belgium
| |
Collapse
|
2
|
Wang M, Czernik PJ, Lecka-Czernik B, Xu Y, Hill JW. IGF-1 and insulin receptors in LepRb neurons jointly regulate body growth, bone mass, reproduction, and metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.20.614140. [PMID: 39345425 PMCID: PMC11429997 DOI: 10.1101/2024.09.20.614140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Leptin receptor (LepRb)-expressing neurons are known to link body growth and reproduction, but whether these functions are mediated via insulin-like growth factor 1 receptor (IGF1R) signaling is unknown. IGF-1 and insulin can bind to each other's receptors, permitting IGF-1 signaling in the absence of IGF1R. Therefore, we created mice lacking IGF1R exclusively in LepRb neurons (IGF1RLepRb mice) and simultaneously lacking IGF1R and insulin receptor (IR) in LepRb neurons (IGF1R/IRLepRb mice) and then characterized their body growth, bone morphology, reproductive and metabolic functions. We found that IGF1R and IR in LepRb neurons were required for normal timing of pubertal onset, while IGF1R in LepRb neurons played a predominant role in regulating adult fertility and exerted protective effects against reproductive aging. Accompanying these reproductive deficits, IGF1RLepRb mice and IGF1R/IRLepRb mice had transient growth retardation. Notably, IGF1R in LepRb neurons was indispensable for normal trabecular and cortical bone mass accrual in both sexes. These findings suggest that IGF1R in LepRb neurons is involved in the interaction among body growth, bone development, and reproduction. Though only mild changes in body weight were detected, simultaneous deletion of IGF1R and IR in LepRb neurons caused dramatically increased fat mass composition, decreased lean mass composition, lower energy expenditure, and locomotor activity in both sexes. Male IGF1R/IRLepRb mice exhibited impaired insulin sensitivity. These findings suggest that IGF1R and IR in LepRb neurons jointly regulated body composition, energy balance, and glucose homeostasis. Taken together, our studies identified the sex-dependent complex roles of IGF1R and IR in LepRb neurons in regulating body growth, reproduction, and metabolism.
Collapse
Affiliation(s)
- Mengjie Wang
- Center for Diabetes and Endocrine Research, Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, Ohio, USA
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Piotr J Czernik
- Center for Diabetes and Endocrine Research, Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, Ohio, USA
| | - Beata Lecka-Czernik
- Center for Diabetes and Endocrine Research, Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, Ohio, USA
- Department of Orthopedic Surgery, University of Toledo College of Medicine, Toledo, Ohio, USA
| | - Yong Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Jennifer W Hill
- Center for Diabetes and Endocrine Research, Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, Ohio, USA
- Department of Obstetrics and Gynecology, University of Toledo College of Medicine, Toledo, Ohio, USA
| |
Collapse
|
3
|
Kauffman AS. Androgen Inhibition of Reproductive Neuroendocrine Function in Females and Transgender Males. Endocrinology 2024; 165:bqae113. [PMID: 39207217 PMCID: PMC11393496 DOI: 10.1210/endocr/bqae113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Ovarian function is controlled by pituitary secretion of luteinizing hormone (LH) and follicle stimulating hormone (FSH), which in turn are governed by gonadotropin releasing hormone (GnRH) secreted from the brain. A fundamental principle of reproductive axis regulation is negative feedback signaling by gonadal sex steroids back to the brain to fine-tune GnRH and gonadotropin secretion. Endogenous negative feedback effects can be mimicked by exogenous steroid treatments, including androgens, in both sexes. Indeed, a growing number of clinical and animal studies indicate that high levels of exogenous androgens, in the typically male physiological range, can inhibit LH secretion in females, as occurs in males. However, the mechanisms by which male-level androgens inhibit GnRH and LH secretion still remain poorly understood, and this knowledge gap is particularly pronounced in transgender men (individuals designated female at birth but identifying as male). Indeed, many transgender men take long-term gender-affirming hormone therapy that mimics male-level testosterone levels. The impact of such gender-affirming testosterone on the reproductive axis, both at the ovarian and neuroendocrine level, is a long-understudied area that still requires further investigation. Importantly, the few concepts of androgen actions in females mostly come from studies of polycystic ovary syndrome, which does not recapitulate a similar androgen milieu or a pathophysiology of inhibited LH secretion as occurs in testosterone-treated transgender men. This review summarizes clinical evidence indicating that exogenous androgens can impair neuroendocrine reproductive function in both female individuals and transgender men and highlights emerging experimental data supporting this in recently developed transgender rodent models.
Collapse
Affiliation(s)
- Alexander S Kauffman
- Department of OBGYN and Reproductive Sciences, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
4
|
Babey ME, Krause WC, Chen K, Herber CB, Torok Z, Nikkanen J, Rodriguez R, Zhang X, Castro-Navarro F, Wang Y, Wheeler EE, Villeda S, Leach JK, Lane NE, Scheller EL, Chan CKF, Ambrosi TH, Ingraham HA. A maternal brain hormone that builds bone. Nature 2024; 632:357-365. [PMID: 38987585 PMCID: PMC11306098 DOI: 10.1038/s41586-024-07634-3] [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: 09/16/2023] [Accepted: 05/30/2024] [Indexed: 07/12/2024]
Abstract
In lactating mothers, the high calcium (Ca2+) demand for milk production triggers significant bone loss1. Although oestrogen normally counteracts excessive bone resorption by promoting bone formation, this sex steroid drops precipitously during this postpartum period. Here we report that brain-derived cellular communication network factor 3 (CCN3) secreted from KISS1 neurons of the arcuate nucleus (ARCKISS1) fills this void and functions as a potent osteoanabolic factor to build bone in lactating females. We began by showing that our previously reported female-specific, dense bone phenotype2 originates from a humoral factor that promotes bone mass and acts on skeletal stem cells to increase their frequency and osteochondrogenic potential. This circulatory factor was then identified as CCN3, a brain-derived hormone from ARCKISS1 neurons that is able to stimulate mouse and human skeletal stem cell activity, increase bone remodelling and accelerate fracture repair in young and old mice of both sexes. The role of CCN3 in normal female physiology was revealed after detecting a burst of CCN3 expression in ARCKISS1 neurons coincident with lactation. After reducing CCN3 in ARCKISS1 neurons, lactating mothers lost bone and failed to sustain their progeny when challenged with a low-calcium diet. Our findings establish CCN3 as a potentially new therapeutic osteoanabolic hormone for both sexes and define a new maternal brain hormone for ensuring species survival in mammals.
Collapse
Affiliation(s)
- Muriel E Babey
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Francisco, San Francisco, CA, USA
| | - William C Krause
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - Kun Chen
- Department of Orthopaedic Surgery, University of California, Davis, Sacramento, CA, USA
| | - Candice B Herber
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
- Denali Therapeutics, South San Francisco, CA, USA
| | - Zsofia Torok
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - Joni Nikkanen
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Ruben Rodriguez
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
- Carmot Therapeutics, Berkeley, CA, USA
| | - Xiao Zhang
- Department of Medicine, Washington University, St Louis, MO, USA
| | - Fernanda Castro-Navarro
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - Yuting Wang
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Erika E Wheeler
- Department of Orthopaedic Surgery, University of California, Davis, Sacramento, CA, USA
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA
| | - Saul Villeda
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
| | - J Kent Leach
- Department of Orthopaedic Surgery, University of California, Davis, Sacramento, CA, USA
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA
| | - Nancy E Lane
- Department of Medicine, Division of Rheumatology, University of California, Davis, Sacramento, CA, USA
| | - Erica L Scheller
- Department of Medicine, Washington University, St Louis, MO, USA
| | - Charles K F Chan
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Thomas H Ambrosi
- Department of Orthopaedic Surgery, University of California, Davis, Sacramento, CA, USA.
| | - Holly A Ingraham
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
5
|
Sáenz de Miera C, Bellefontaine N, Allen SJ, Myers MG, Elias CF. Glutamate neurotransmission from leptin receptor cells is required for typical puberty and reproductive function in female mice. eLife 2024; 13:RP93204. [PMID: 39007235 PMCID: PMC11249761 DOI: 10.7554/elife.93204] [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] [Indexed: 07/16/2024] Open
Abstract
The hypothalamic ventral premammillary nucleus (PMv) is a glutamatergic nucleus essential for the metabolic control of reproduction. However, conditional deletion of leptin receptor long form (LepRb) in vesicular glutamate transporter 2 (Vglut2) expressing neurons results in virtually no reproductive deficits. In this study, we determined the role of glutamatergic neurotransmission from leptin responsive PMv neurons on puberty and fertility. We first assessed if stimulation of PMv neurons induces luteinizing hormone (LH) release in fed adult females. We used the stimulatory form of designer receptor exclusively activated by designer drugs (DREADDs) in LeprCre (LepRb-Cre) mice. We collected blood sequentially before and for 1 hr after intravenous clozapine-N-oxide injection. LH level increased in animals correctly targeted to the PMv, and LH level was correlated to the number of Fos immunoreactive neurons in the PMv. Next, females with deletion of Slc17a6 (Vglut2) in LepRb neurons (LeprΔVGlut2) showed delayed age of puberty, disrupted estrous cycles, increased gonadotropin-releasing hormone (GnRH) concentration in the axon terminals, and disrupted LH secretion, suggesting impaired GnRH release. To assess if glutamate is required for PMv actions in pubertal development, we generated a Cre-induced reexpression of endogenous LepRb (LeprloxTB) with concomitant deletion of Slc17a6 (Vglut2flox) mice. Rescue of Lepr and deletion of Slc17a6 in the PMv was obtained by stereotaxic injection of an adeno-associated virus vector expressing Cre recombinase. Control LeprloxTB mice with PMv LepRb rescue showed vaginal opening, follicle maturation, and became pregnant, while LeprloxTB;Vglut2flox mice showed no pubertal development. Our results indicate that glutamatergic neurotransmission from leptin sensitive neurons regulates the reproductive axis, and that leptin action on pubertal development via PMv neurons requires Vglut2.
Collapse
Affiliation(s)
- Cristina Sáenz de Miera
- Department of Molecular and Integrative Physiology, University of Michigan–Ann ArborAnn ArborUnited States
| | - Nicole Bellefontaine
- Department of Molecular and Integrative Physiology, University of Michigan–Ann ArborAnn ArborUnited States
| | - Susan J Allen
- Department of Molecular and Integrative Physiology, University of Michigan–Ann ArborAnn ArborUnited States
| | - Martin G Myers
- Department of Molecular and Integrative Physiology, University of Michigan–Ann ArborAnn ArborUnited States
- Elizabeth W. Caswell Diabetes Institute, University of Michigan–Ann ArborAnn ArborUnited States
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan–Ann ArborAnn ArborUnited States
| | - Carol F Elias
- Department of Molecular and Integrative Physiology, University of Michigan–Ann ArborAnn ArborUnited States
- Elizabeth W. Caswell Diabetes Institute, University of Michigan–Ann ArborAnn ArborUnited States
- Department of Obstetrics and Gynecology, University of Michigan–Ann ArborAnn ArborUnited States
| |
Collapse
|
6
|
Ow MC, Nishiguchi MA, Dar AR, Butcher RA, Hall SE. RNAi-dependent expression of sperm genes in ADL chemosensory neurons is required for olfactory responses in Caenorhabditis elegans. Front Mol Biosci 2024; 11:1396587. [PMID: 39055986 PMCID: PMC11269235 DOI: 10.3389/fmolb.2024.1396587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 06/11/2024] [Indexed: 07/28/2024] Open
Abstract
Environmental conditions experienced early in the life of an animal can result in gene expression changes later in its life history. We have previously shown that C. elegans animals that experienced the developmentally arrested and stress resistant dauer stage (postdauers) retain a cellular memory of early-life stress that manifests during adulthood as genome-wide changes in gene expression, chromatin states, and altered life history traits. One consequence of developmental reprogramming in C. elegans postdauer adults is the downregulation of osm-9 TRPV channel gene expression in the ADL chemosensory neurons resulting in reduced avoidance to a pheromone component, ascr#3. This altered response to ascr#3 requires the principal effector of the somatic nuclear RNAi pathway, the Argonaute (AGO) NRDE-3. To investigate the role of the somatic nuclear RNAi pathway in regulating the developmental reprogramming of ADL due to early-life stress, we profiled the mRNA transcriptome of control and postdauer ADL in wild-type and nrde-3 mutant adults. We found 711 differentially expressed (DE) genes between control and postdauer ADL neurons, 90% of which are dependent upon NRDE-3. Additionally, we identified a conserved sequence that is enriched in the upstream regulatory sequences of the NRDE-3-dependent differentially expressed genes. Surprisingly, 214 of the ADL DE genes are considered "germline-expressed", including 21 genes encoding the Major Sperm Proteins and two genes encoding the sperm-specific PP1 phosphatases, GSP-3 and GSP-4. Loss of function mutations in gsp-3 resulted in both aberrant avoidance and attraction behaviors. We also show that an AGO pseudogene, Y49F6A.1 (wago-11), is expressed in ADL and is required for ascr#3 avoidance. Overall, our results suggest that small RNAs and reproductive genes program the ADL mRNA transcriptome during their developmental history and highlight a nexus between neuronal and reproductive networks in calibrating animal neuroplasticity.
Collapse
Affiliation(s)
- Maria C. Ow
- Biology Department, Syracuse University, Syracuse, NY, United States
| | | | - Abdul Rouf Dar
- Department of Chemistry, University of Florida, Gainesville, FL, United States
| | - Rebecca A. Butcher
- Department of Chemistry, University of Florida, Gainesville, FL, United States
| | - Sarah E. Hall
- Biology Department, Syracuse University, Syracuse, NY, United States
| |
Collapse
|
7
|
Torres T, Adam N, Mhaouty-Kodja S, Naulé L. Reproductive function and behaviors: an update on the role of neural estrogen receptors alpha and beta. Front Endocrinol (Lausanne) 2024; 15:1408677. [PMID: 38978624 PMCID: PMC11228153 DOI: 10.3389/fendo.2024.1408677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 05/29/2024] [Indexed: 07/10/2024] Open
Abstract
Infertility is becoming a major public health problem, with increasing frequency due to medical, environmental and societal causes. The increasingly late age of childbearing, growing exposure to endocrine disruptors and other reprotoxic products, and increasing number of medical reproductive dysfunctions (endometriosis, polycystic ovary syndrome, etc.) are among the most common causes. Fertility relies on fine-tuned control of both neuroendocrine function and reproductive behaviors, those are critically regulated by sex steroid hormones. Testosterone and estradiol exert organizational and activational effects throughout life to establish and activate the neural circuits underlying reproductive function. This regulation is mediated through estrogen receptors (ERs) and androgen receptor (AR). Estradiol acts mainly via nuclear estrogen receptors ERα and ERβ. The aim of this review is to summarize the genetic studies that have been undertaken to comprehend the specific contribution of ERα and ERβ in the neural circuits underlying the regulation of the hypothalamic-pituitary-gonadal axis and the expression of reproductive behaviors, including sexual and parental behavior. Particular emphasis will be placed on the neural role of these receptors and the underlying sex differences.
Collapse
Affiliation(s)
| | | | | | - Lydie Naulé
- Sorbonne Université, CNRS UMR8246, INSERM U1130, Neuroscience Paris Seine – Institut de Biologie Paris Seine, Paris, France
| |
Collapse
|
8
|
Szabó F, Köves K, Gál L. History of the Development of Knowledge about the Neuroendocrine Control of Ovulation-Recent Knowledge on the Molecular Background. Int J Mol Sci 2024; 25:6531. [PMID: 38928237 PMCID: PMC11203711 DOI: 10.3390/ijms25126531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/30/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
The physiology of reproduction has been of interest to researchers for centuries. The purpose of this work is to review the development of our knowledge on the neuroendocrine background of the regulation of ovulation. We first describe the development of the pituitary gland, the structure of the median eminence (ME), the connection between the hypothalamus and the pituitary gland, the ovarian and pituitary hormones involved in ovulation, and the pituitary cell composition. We recall the pioneer physiological and morphological investigations that drove development forward. The description of the supraoptic-paraventricular magnocellular and tuberoinfundibular parvocellular systems and recognizing the role of the hypophysiotropic area were major milestones in understanding the anatomical and physiological basis of reproduction. The discovery of releasing and inhibiting hormones, the significance of pulse and surge generators, the pulsatile secretion of the gonadotropin-releasing hormone (GnRH), and the subsequent pulsatility of luteinizing (LH) and follicle-stimulating hormones (FSH) in the human reproductive physiology were truly transformative. The roles of three critical neuropeptides, kisspeptin (KP), neurokinin B (NKB), and dynorphin (Dy), were also identified. This review also touches on the endocrine background of human infertility and assisted fertilization.
Collapse
Affiliation(s)
- Flóra Szabó
- Division of Gastroenterology and Nutrition, Children’s Hospital of Richmond, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Katalin Köves
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, 1094 Budapest, Hungary
| | - Levente Gál
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA 24061, USA;
| |
Collapse
|
9
|
de Miera CS, Bellefontaine N, Allen SJ, Myers MG, Elias CF. Glutamate neurotransmission from leptin receptor cells is required for typical puberty and reproductive function in female mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.21.558865. [PMID: 37790549 PMCID: PMC10542178 DOI: 10.1101/2023.09.21.558865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
The hypothalamic ventral premammillary nucleus (PMv) is a glutamatergic nucleus essential for the metabolic control of reproduction. However, conditional deletion of leptin receptor (LepRb) in vesicular glutamate transporter 2 (Vglut2) expressing neurons results in virtually no reproductive deficits. In this study, we determine the role of glutamatergic signaling from leptin responsive PMv neurons on puberty and fertility. We first assessed if stimulation of PMv neurons induces LH release in fed adult females. We used the stimulatory form of designer receptor exclusively activated by designer drugs (DREADDs) in LepRb-Cre mice. We collected blood sequentially before and for 1h after iv. clozapine-N-oxide injection. LH level increased in animals correctly targeted to the PMv, and LH level was correlated to the number of cFos immunoreactive neurons in the PMv. Next, females with deletion of Vglut2 in LepRb neurons (LepR∆VGlut2) showed delayed age of puberty, disrupted estrous cycles, increased GnRH concentration in the axon terminals and disrupted LH responses, suggesting impaired GnRH release. To assess if glutamate is required for PMv actions in pubertal development, we generated a Cre-induced reexpression of endogenous LepRb (LepRloxTB) with concomitant deletion of Vglut2 (Vglut2-floxed) mice. Rescue of Lepr and deletion of Vglut2 in the PMv was obtained by stereotaxic injection of an adeno-associated virus vector expressing Cre recombinase. Control LepRloxTB mice with PMv LepRb rescue showed vaginal opening, follicle maturation and became pregnant, while LepRloxTB;Vglut2flox mice showed no pubertal development. Our results indicate that glutamatergic signaling from leptin sensitive neurons regulates the reproductive axis, and that leptin action on pubertal development via PMv neurons requires Vglut2.
Collapse
Affiliation(s)
- Cristina Sáenz de Miera
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109-5622, USA
| | - Nicole Bellefontaine
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109-5622, USA
| | - Susan J. Allen
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109-5622, USA
| | - Martin G. Myers
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109-5622, USA
- Elizabeth W. Caswell Diabetes Institute, University of Michigan, Ann Arbor, MI, 48109-5622, USA
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI, 48109-5622, USA
| | - Carol F. Elias
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109-5622, USA
- Elizabeth W. Caswell Diabetes Institute, University of Michigan, Ann Arbor, MI, 48109-5622, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, 48109-5622, USA
| |
Collapse
|
10
|
Poitras M, Morin A, Bentley GE, Plamondon H. Global cerebral ischemia in adult female rats interrupts estrous cyclicity and induces lasting changes in hypothalamic-pituitary-gonadal axis signaling peptides. Neurosci Lett 2024; 819:137578. [PMID: 38048875 DOI: 10.1016/j.neulet.2023.137578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/20/2023] [Accepted: 12/01/2023] [Indexed: 12/06/2023]
Abstract
Persistent post-ischemic alterations to the hypothalamic-pituitary-adrenal (HPA) axis occur following global cerebral ischemia (GCI) in rodents. However, similar effects on hypothalamic-pituitary-gonadal (HPG) axis activation remain to be determined. Therefore, this study evaluated the effects of GCI in adult female rats (via four-vessel occlusion) on the regularity of the estrous cycle for 24-days post ischemia. A second objective aimed to assess persistent alterations of HPG axis activation through determination of the expression of estrogen receptor alpha (ERα), kisspeptin (Kiss1), and gonadotropin-inhibitory hormone (GnIH/RFamide-related peptide; RFRP3) in the medial preoptic area (POA), arcuate nucleus (ARC), dorsomedial nucleus (DMH) of the hypothalamus, and CA1 of the hippocampus 25 days post ischemia. Expression of glucocorticoid receptors (GR) in the paraventricular nucleus of the hypothalamus (PVN) and CA1 served as a proxy of altered HPA axis activation. Our findings demonstrated interruption of the estrous cycle in 87.5 % of ischemic rats, marked by persistent diestrus, lasting on average 11.86 days. Moreover, compared to sham-operated controls, ischemic female rats showed reduced Kiss1 expression in the hypothalamic ARC and POA, concomitant with elevated ERα in the ARC and increased GnIH in the DMH and CA1. Reduced GR expression in the CA1 was associated with increased GR-immunoreactivity in the PVN, indicative of lasting dysregulation of HPA axis activation. Together, these findings demonstrate GCI disruption of female rats' estrous cycle over multiple days, with a lasting impact on HPG axis regulators within the reproductive axis.
Collapse
Affiliation(s)
- Marilou Poitras
- Cerebro Vascular Accidents and Behavioural Recovery Laboratory, School of Psychology, University of Ottawa, Ottawa, Canada
| | - Alexandre Morin
- Cerebro Vascular Accidents and Behavioural Recovery Laboratory, School of Psychology, University of Ottawa, Ottawa, Canada
| | - George E Bentley
- Department of Integrative Biology, University of California, Berkeley, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Hélène Plamondon
- Cerebro Vascular Accidents and Behavioural Recovery Laboratory, School of Psychology, University of Ottawa, Ottawa, Canada.
| |
Collapse
|
11
|
Clarkson J, Yip SH, Porteous R, Kauff A, Heather AK, Herbison AE. CRISPR-Cas9 knockdown of ESR1 in preoptic GABA-kisspeptin neurons suppresses the preovulatory surge and estrous cycles in female mice. eLife 2023; 12:RP90959. [PMID: 38126277 PMCID: PMC10735218 DOI: 10.7554/elife.90959] [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] [Indexed: 12/23/2023] Open
Abstract
Evidence suggests that estradiol-sensing preoptic area GABA neurons are involved in the preovulatory surge mechanism necessary for ovulation. In vivo CRISPR-Cas9 editing was used to achieve a 60-70% knockdown in estrogen receptor alpha (ESR1) expression by GABA neurons located within the regions of the rostral periventricular area of the third ventricle (RP3V) and medial preoptic nuclei (MPN) in adult female mice. Mice exhibited variable reproductive phenotypes with the only significant finding being mice with bilateral ESR1 deletion in RP3V GABA neurons having reduced cFos expression in gonadotropin-releasing hormone (GnRH) neurons at the time of the surge. One sub-population of RP3V GABA neurons expresses kisspeptin. Re-grouping ESR1-edited mice on the basis of their RP3V kisspeptin expression revealed a highly consistent phenotype; mice with a near-complete loss of kisspeptin immunoreactivity displayed constant estrus and failed to exhibit surge activation but retained pulsatile luteinizing hormone (LH) secretion. These observations demonstrate that ESR1-expressing GABA-kisspeptin neurons in the RP3V are essential for the murine preovulatory LH surge mechanism.
Collapse
Affiliation(s)
- Jenny Clarkson
- Centre for NeuroendocrinologyDunedinNew Zealand
- Department of Physiology, University of Otago School of Biomedical SciencesDunedinNew Zealand
| | - Siew Hoong Yip
- Centre for NeuroendocrinologyDunedinNew Zealand
- Department of Physiology, University of Otago School of Biomedical SciencesDunedinNew Zealand
| | - Robert Porteous
- Centre for NeuroendocrinologyDunedinNew Zealand
- Department of Physiology, University of Otago School of Biomedical SciencesDunedinNew Zealand
| | - Alexia Kauff
- Department of Physiology, University of Otago School of Biomedical SciencesDunedinNew Zealand
| | - Alison K Heather
- Department of Physiology, University of Otago School of Biomedical SciencesDunedinNew Zealand
| | - Allan E Herbison
- Centre for NeuroendocrinologyDunedinNew Zealand
- Department of Physiology, University of Otago School of Biomedical SciencesDunedinNew Zealand
- Department of Physiology, Development and Neuroscience, University of CambridgeCambridgeUnited Kingdom
| |
Collapse
|
12
|
Cutia CA, Christian-Hinman CA. Mechanisms linking neurological disorders with reproductive endocrine dysfunction: Insights from epilepsy research. Front Neuroendocrinol 2023; 71:101084. [PMID: 37506886 PMCID: PMC10818027 DOI: 10.1016/j.yfrne.2023.101084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 07/03/2023] [Accepted: 07/25/2023] [Indexed: 07/30/2023]
Abstract
Gonadal hormone actions in the brain can both worsen and alleviate symptoms of neurological disorders. Although neurological conditions and reproductive endocrine function are seemingly disparate, compelling evidence indicates that reciprocal interactions exist between certain disorders and hypothalamic-pituitary-gonadal (HPG) axis irregularities. Epilepsy is a neurological disorder that shows significant reproductive endocrine dysfunction (RED) in clinical populations. Seizures, particularly those arising from temporal lobe structures, can drive HPG axis alterations, and hormones produced in the HPG axis can reciprocally modulate seizure activity. Despite this relationship, mechanistic links between seizures and RED, and vice versa, are still largely unknown. Here, we review clinical evidence alongside recent investigations in preclinical animal models into the contributions of seizures to HPG axis malfunction, describe the effects of HPG axis hormonal feedback on seizure activity, and discuss how epilepsy research can offer insight into mechanisms linking neurological disorders to HPG axis dysfunction, an understudied area of neuroendocrinology.
Collapse
Affiliation(s)
- Cathryn A Cutia
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Catherine A Christian-Hinman
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, USA; Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL, USA; Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
13
|
Sáenz de Miera C, Feng J, Elias CF, Qi N. Remote Neuronal Activation Coupled with Automated Blood Sampling to Induce and Measure Circulating Luteinizing Hormone in Mice. J Vis Exp 2023:10.3791/65875. [PMID: 37677024 PMCID: PMC11026941 DOI: 10.3791/65875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2023] Open
Abstract
Circulating luteinizing hormone (LH) levels are an essential index of the functioning of the hypothalamic-pituitary control of reproduction. The role of numerous inputs and neuronal populations in the modulation of LH release is still unknown. Measuring changes in LH levels in mice is often a challenge since they are easily disrupted by environmental stress. Current techniques to measure LH release and pulsatility require long-term training for mice to adapt to manipulation stress, certain restraint, the presence of the investigator, and working on individual animals, reducing its usefulness for many research questions. This paper presents a technique to remotely activate specific neuronal populations using Designer Receptor Exclusively Activated by Designer Drugs (DREADDs) technology coupled with automated sequential blood sampling in conscious, freely moving, and undisturbed mice. We first describe the stereotaxic surgery protocol to deliver adeno-associated virus (AAV) vectors expressing DREADDs to specific neuronal populations. Next, we describe the protocol for carotid artery and jugular vein cannulation and postsurgical connection to the CULEX automated blood sampling system. Finally, we describe the protocol for clozapine-N-oxide intravenous injection for remote neuronal activation and automated blood collection. This technique allows for programmed automated sampling every 5 min or longer for a given period, coupled with intravenous substance injection at a desired time point or duration. Overall, we found this technique to be a powerful approach for research on neuroendocrine control.
Collapse
Affiliation(s)
| | - Jiane Feng
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor; Mouse Metabolic Phenotyping Center-Live, University of Michigan, Ann Arbor
| | - Carol F Elias
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor; Mouse Metabolic Phenotyping Center-Live, University of Michigan, Ann Arbor
| | - Nathan Qi
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor; Mouse Metabolic Phenotyping Center-Live, University of Michigan, Ann Arbor;
| |
Collapse
|
14
|
Isola JVV, Ko S, Ocañas SR, Stout MB. Role of Estrogen Receptor α in Aging and Chronic Disease. ADVANCES IN GERIATRIC MEDICINE AND RESEARCH 2023; 5:e230005. [PMID: 37425648 PMCID: PMC10327608 DOI: 10.20900/agmr20230005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Estrogen receptor alpha (ERα) plays a crucial role in reproductive function in both sexes. It also mediates cellular responses to estrogens in multiple nonreproductive organ systems, many of which regulate systemic metabolic homeostasis and inflammatory processes in mammals. The loss of estrogens and/or ERα agonism during aging is associated with the emergence of several comorbid conditions, particularly in females undergoing the menopausal transition. Emerging data also suggests that male mammals likely benefit from ERα agonism if done in a way that circumvents feminizing characteristics. This has led us, and others, to speculate that tissue-specific ERα agonism may hold therapeutic potential for curtailing aging and chronic disease burden in males and females that are at high-risk of cancer and/or cardiovascular events with traditional estrogen replacement therapies. In this mini-review, we emphasize the role of ERα in the brain and liver, summarizing recent evidence that indicates these two organs systems mediate the beneficial effects of estrogens on metabolism and inflammation during aging. We also discuss how 17α-estradiol administration elicits health benefits in an ERα-dependent manner, which provides proof-of-concept that ERα may be a druggable target for attenuating aging and age-related disease burden.
Collapse
Affiliation(s)
- José V. V. Isola
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Sunghwan Ko
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Sarah R. Ocañas
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA
| | - Michael B. Stout
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
15
|
Ramos-Pittol JM, Fernandes-Freitas I, Milona A, Manchishi SM, Rainbow K, Lam BYH, Tadross JA, Beucher A, Colledge WH, Cebola I, Murphy KG, Miguel-Aliaga I, Yeo GSH, Dhillo WS, Owen BM. Dax1 modulates ERα-dependent hypothalamic estrogen sensing in female mice. Nat Commun 2023; 14:3076. [PMID: 37248237 PMCID: PMC10227040 DOI: 10.1038/s41467-023-38618-y] [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: 05/23/2022] [Accepted: 05/10/2023] [Indexed: 05/31/2023] Open
Abstract
Coupling the release of pituitary hormones to the developmental stage of the oocyte is essential for female fertility. It requires estrogen to restrain kisspeptin (KISS1)-neuron pulsatility in the arcuate hypothalamic nucleus, while also exerting a surge-like effect on KISS1-neuron activity in the AVPV hypothalamic nucleus. However, a mechanistic basis for this region-specific effect has remained elusive. Our genomic analysis in female mice demonstrate that some processes, such as restraint of KISS1-neuron activity in the arcuate nucleus, may be explained by region-specific estrogen receptor alpha (ERα) DNA binding at gene regulatory regions. Furthermore, we find that the Kiss1-locus is uniquely regulated in these hypothalamic nuclei, and that the nuclear receptor co-repressor NR0B1 (DAX1) restrains its transcription specifically in the arcuate nucleus. These studies provide mechanistic insight into how ERα may control the KISS1-neuron, and Kiss1 gene expression, to couple gonadotropin release to the developmental stage of the oocyte.
Collapse
Affiliation(s)
- Jose M Ramos-Pittol
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, 6020, Austria
| | | | - Alexandra Milona
- Institute of Clinical Sciences, Imperial College London, London, United Kingdom
| | - Stephen M Manchishi
- Department of Physiology, Development, and Neuroscience, Cambridge University, Cambridge, United Kingdom
| | - Kara Rainbow
- Medical Research Council Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science-Metabolic Research Laboratories, Cambridge University, Cambridge, United Kingdom
| | - Brian Y H Lam
- Medical Research Council Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science-Metabolic Research Laboratories, Cambridge University, Cambridge, United Kingdom
| | - John A Tadross
- Medical Research Council Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science-Metabolic Research Laboratories, Cambridge University, Cambridge, United Kingdom
- Department of Histopathology and East Midlands & East of England Genomic Laboratory Hub, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Anthony Beucher
- Section of Genetics and Genomics, Imperial College London, London, United Kingdom
| | - William H Colledge
- Department of Physiology, Development, and Neuroscience, Cambridge University, Cambridge, United Kingdom
| | - Inês Cebola
- Section of Genetics and Genomics, Imperial College London, London, United Kingdom
| | - Kevin G Murphy
- Section of Investigative Medicine, Imperial College London, London, United Kingdom
| | - Irene Miguel-Aliaga
- Institute of Clinical Sciences, Imperial College London, London, United Kingdom
- MRC London Institute of Medical Sciences, London, United Kingdom
| | - Giles S H Yeo
- Medical Research Council Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science-Metabolic Research Laboratories, Cambridge University, Cambridge, United Kingdom
| | - Waljit S Dhillo
- Section of Investigative Medicine, Imperial College London, London, United Kingdom.
| | - Bryn M Owen
- Section of Investigative Medicine, Imperial College London, London, United Kingdom.
| |
Collapse
|
16
|
Starrett JR, Moenter SM. Hypothalamic kisspeptin neurons as potential mediators of estradiol negative and positive feedback. Peptides 2023; 163:170963. [PMID: 36740189 PMCID: PMC10516609 DOI: 10.1016/j.peptides.2023.170963] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/09/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
Gonadal steroid feedback regulates the brain's patterned secretion of gonadotropin-releasing hormone (GnRH). Negative feedback, which occurs in males and during the majority of the female cycle, modulates the amplitude and frequency of GnRH pulses. Positive feedback occurs in females when high estradiol induces a surge pattern of GnRH release. These two forms of feedback and their corresponding patterns of GnRH secretion are thought to be mediated by kisspeptin-expressing neurons in two hypothalamic areas: the arcuate nucleus and the anteroventral periventricular area. In this review, we present evidence for this theory and remaining questions to be addressed.
Collapse
Affiliation(s)
- J Rudolph Starrett
- Departments of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Suzanne M Moenter
- Departments of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA; Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA; Obstetrics & Gynecology, University of Michigan, Ann Arbor, MI 48109, USA; The Reproductive Sciences Program, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
17
|
McQuillan HJ, Clarkson J, Kauff A, Han SY, Yip SH, Cheong I, Porteous R, Heather AK, Herbison AE. Definition of the estrogen negative feedback pathway controlling the GnRH pulse generator in female mice. Nat Commun 2022; 13:7433. [PMID: 36460649 PMCID: PMC9718805 DOI: 10.1038/s41467-022-35243-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 11/23/2022] [Indexed: 12/03/2022] Open
Abstract
The mechanisms underlying the homeostatic estrogen negative feedback pathway central to mammalian fertility have remained unresolved. Direct measurement of gonadotropin-releasing hormone (GnRH) pulse generator activity in freely behaving mice with GCaMP photometry demonstrated striking estradiol-dependent plasticity in the frequency, duration, amplitude, and profile of pulse generator synchronization events. Mice with Cre-dependent deletion of ESR1 from all kisspeptin neurons exhibited pulse generator activity identical to that of ovariectomized wild-type mice. An in vivo CRISPR-Cas9 approach was used to knockdown ESR1 expression selectively in arcuate nucleus (ARN) kisspeptin neurons. Mice with >80% deletion of ESR1 in ARN kisspeptin neurons exhibited the ovariectomized pattern of GnRH pulse generator activity and high frequency LH pulses but with very low amplitude due to reduced responsiveness of the pituitary. Together, these studies demonstrate that estrogen utilizes ESR1 in ARN kisspeptin neurons to achieve estrogen negative feedback of the GnRH pulse generator in mice.
Collapse
Affiliation(s)
- H James McQuillan
- Centre for Neuroendocrinology, University of Otago School of Biomedical Sciences, Dunedin, 9054, New Zealand.,Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, 9054, New Zealand
| | - Jenny Clarkson
- Centre for Neuroendocrinology, University of Otago School of Biomedical Sciences, Dunedin, 9054, New Zealand.,Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, 9054, New Zealand
| | - Alexia Kauff
- Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, 9054, New Zealand
| | - Su Young Han
- Centre for Neuroendocrinology, University of Otago School of Biomedical Sciences, Dunedin, 9054, New Zealand.,Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, 9054, New Zealand.,Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EG, UK
| | - Siew Hoong Yip
- Centre for Neuroendocrinology, University of Otago School of Biomedical Sciences, Dunedin, 9054, New Zealand.,Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, 9054, New Zealand
| | - Isaiah Cheong
- Centre for Neuroendocrinology, University of Otago School of Biomedical Sciences, Dunedin, 9054, New Zealand.,Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, 9054, New Zealand
| | - Robert Porteous
- Centre for Neuroendocrinology, University of Otago School of Biomedical Sciences, Dunedin, 9054, New Zealand.,Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, 9054, New Zealand
| | - Alison K Heather
- Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, 9054, New Zealand
| | - Allan E Herbison
- Centre for Neuroendocrinology, University of Otago School of Biomedical Sciences, Dunedin, 9054, New Zealand. .,Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, 9054, New Zealand. .,Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EG, UK.
| |
Collapse
|
18
|
Constantin S. Targeting KNDy neurons to control GnRH pulses. Curr Opin Pharmacol 2022; 67:102316. [PMID: 36347163 PMCID: PMC9772270 DOI: 10.1016/j.coph.2022.102316] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/03/2022] [Accepted: 10/07/2022] [Indexed: 11/06/2022]
Abstract
Gonadotropin-releasing hormone (GnRH) is the final output of the central nervous system that drives fertility. A characteristic of GnRH secretion is its pulsatility, which is driven by a pulse generator. Each GnRH pulse triggers a luteinizing hormone (LH) pulse. However, the puzzle has been to reconcile the synchronicity of GnRH neurons with the scattered hypothalamic distribution of their cell bodies. A leap toward understanding GnRH pulses was the discovery of kisspeptin neurons near the distal processes of GnRH neurons, which secrete kisspeptins, potent excitatory neuropeptides on GnRH neurons, and equipped with dual, but opposite, self-modulatory neuropeptides, neurokinin B and dynorphin. Over the last decade, this cell-to-cell communication has been dissected in animal models. Today the 50-year quest for the basic mechanism of GnRH pulse generation may be over, but questions about its physiological tuning remain. Here is an overview of recent basic research that frames translational research.
Collapse
Affiliation(s)
- Stephanie Constantin
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
19
|
Campideli-Santana AC, Gusmao DO, Almeida FRCL, Araujo-Lopes R, Szawka RE. Partial loss of arcuate kisspeptin neurons in female rats stimulates luteinizing hormone and decreases prolactin secretion induced by estradiol. J Neuroendocrinol 2022; 34:e13204. [PMID: 36319592 DOI: 10.1111/jne.13204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/25/2022] [Accepted: 09/30/2022] [Indexed: 11/28/2022]
Abstract
Kisspeptin, neurokinin, and dynorphin (KNDy) neurons in the arcuate nucleus (ARC) control luteinizing hormone (LH) and prolactin (PRL) release, although their role in conveying the effects of estradiol (E2 ) to these hormones is not well understood. We performed a longitudinal evaluation of female rats in which KNDy neurons were ablated using a neurokinin-3 receptor agonist conjugated with saporin (NK3-SAP) to investigate the impact of the reduction of KNDy neurons on the E2 regulation of gonadal and PRL axes. NK3-SAP rats, bearing a moderate loss of ARC kisspeptin-immunoreactive (-IR) neurons (50%-90%), displayed irregular estrous cycles but essentially unaltered follicular development and a normal number of corpora lutea. Rats were then ovariectomized (OVX) and treated with a positive-feedback dose of E2 (OVX + E2 ). LH and PRL were measured in the tail blood by an enzyme-linked immunosorbent assay. The E2 -induced LH surge was amplified, whereas the PRL rise was decreased in NK3-SAP rats compared to Blank-SAP control. After 10 days of no hormonal treatment, basal LH levels were equally elevated in NK3-SAP and controls. Tyrosine hydroxylase (TH) phosphorylation in the median eminence, in turn, was increased in NK3-SAP rats, with no change in the number of ARC TH-IR neurons. Thus, KNDy neurons exert concurrent and opposite roles in the E2 -induced surges of LH and PRL. The partial loss of KNDy neurons disrupts ovarian cyclicity but does not preclude ovulation, consistent with the disinhibition of the LH preovulatory surge. Conversely, KNDy neurons tonically inhibit the enzymatic activity of tuberoinfundibular dopaminergic neurons, which appears to facilitate PRL release in response to E2 .
Collapse
Affiliation(s)
- Ana C Campideli-Santana
- Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Daniela O Gusmao
- Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Fernanda R C L Almeida
- Departamento de Morfologia, Instituto de Ciencias Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Roberta Araujo-Lopes
- Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Raphael E Szawka
- Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
20
|
Ingram RJ, Leverton LK, Daniels VC, Li J, Christian-Hinman CA. Increased GABA transmission to GnRH neurons after intrahippocampal kainic acid injection in mice is sex-specific and associated with estrous cycle disruption. Neurobiol Dis 2022; 172:105822. [PMID: 35868435 PMCID: PMC9455811 DOI: 10.1016/j.nbd.2022.105822] [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: 05/20/2022] [Revised: 06/29/2022] [Accepted: 07/16/2022] [Indexed: 11/25/2022] Open
Abstract
Patients with epilepsy develop reproductive endocrine comorbidities at a rate higher than that of the general population. Clinical studies have identified disrupted luteinizing hormone (LH) release patterns in patients of both sexes, suggesting potential epilepsy-associated changes in hypothalamic gonadotropin-releasing hormone (GnRH) neuron function. In previous work, we found that GnRH neuron firing is increased in diestrous females and males in the intrahippocampal kainic acid (IHKA) mouse model of temporal lobe epilepsy. Notably, GABAA receptor activation is depolarizing in adult GnRH neurons. Therefore, here we tested the hypothesis that increased GnRH neuron firing in IHKA mice is associated with increased GABAergic drive to GnRH neurons. When ionotropic glutamate receptors (iGluRs) were blocked to isolate GABAergic postsynaptic currents (PSCs), no differences in PSC frequency were seen between GnRH neurons from control and IHKA diestrous females. In the absence of iGluR blockade, however, GABA PSC frequency was increased in GnRH neurons from IHKA females with disrupted estrous cycles, but not saline-injected controls nor IHKA females without estrous cycle disruption. GABA PSC amplitude was also increased in IHKA females with disrupted estrous cycles. These findings suggest the presence of an iGluR-dependent increase in feed-forward GABAergic transmission to GnRH neurons specific to IHKA females with comorbid cycle disruption. In males, GABA PSC frequency and amplitude were unchanged but PSC duration was reduced. Together, these findings suggest that increased GABA transmission helps drive elevated firing in IHKA females on diestrus and indicate the presence of a sex-specific hypothalamic mechanism underlying reproductive endocrine dysfunction in IHKA mice.
Collapse
Affiliation(s)
- Robbie J Ingram
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States of America
| | - Leanna K Leverton
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States of America
| | - Victoria C Daniels
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States of America
| | - Jiang Li
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States of America
| | - Catherine A Christian-Hinman
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States of America; Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States of America; Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States of America.
| |
Collapse
|
21
|
Moeller JS, Bever SR, Finn SL, Phumsatitpong C, Browne MF, Kriegsfeld LJ. Circadian Regulation of Hormonal Timing and the Pathophysiology of Circadian Dysregulation. Compr Physiol 2022; 12:4185-4214. [PMID: 36073751 DOI: 10.1002/cphy.c220018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Circadian rhythms are endogenously generated, daily patterns of behavior and physiology that are essential for optimal health and disease prevention. Disruptions to circadian timing are associated with a host of maladies, including metabolic disease and obesity, diabetes, heart disease, cancer, and mental health disturbances. The circadian timing system is hierarchically organized, with a master circadian clock located in the suprachiasmatic nucleus (SCN) of the anterior hypothalamus and subordinate clocks throughout the CNS and periphery. The SCN receives light information via a direct retinal pathway, synchronizing the master clock to environmental time. At the cellular level, circadian rhythms are ubiquitous, with rhythms generated by interlocking, autoregulatory transcription-translation feedback loops. At the level of the SCN, tight cellular coupling maintains rhythms even in the absence of environmental input. The SCN, in turn, communicates timing information via the autonomic nervous system and hormonal signaling. This signaling couples individual cellular oscillators at the tissue level in extra-SCN brain loci and the periphery and synchronizes subordinate clocks to external time. In the modern world, circadian disruption is widespread due to limited exposure to sunlight during the day, exposure to artificial light at night, and widespread use of light-emitting electronic devices, likely contributing to an increase in the prevalence, and the progression, of a host of disease states. The present overview focuses on the circadian control of endocrine secretions, the significance of rhythms within key endocrine axes for typical, homeostatic functioning, and implications for health and disease when dysregulated. © 2022 American Physiological Society. Compr Physiol 12: 1-30, 2022.
Collapse
Affiliation(s)
- Jacob S Moeller
- Graduate Group in Endocrinology, University of California, Berkeley, California, USA
| | - Savannah R Bever
- Department of Psychology, University of California, Berkeley, California, USA
| | - Samantha L Finn
- Department of Psychology, University of California, Berkeley, California, USA
| | | | - Madison F Browne
- Department of Psychology, University of California, Berkeley, California, USA
| | - Lance J Kriegsfeld
- Graduate Group in Endocrinology, University of California, Berkeley, California, USA.,Department of Psychology, University of California, Berkeley, California, USA.,Department of Integrative Biology, University of California, Berkeley, California, USA.,The Helen Wills Neuroscience Institute, University of California, Berkeley, California, USA
| |
Collapse
|
22
|
Kauffman AS. Neuroendocrine mechanisms underlying estrogen positive feedback and the LH surge. Front Neurosci 2022; 16:953252. [PMID: 35968365 PMCID: PMC9364933 DOI: 10.3389/fnins.2022.953252] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/08/2022] [Indexed: 01/26/2023] Open
Abstract
A fundamental principle in reproductive neuroendocrinology is sex steroid feedback: steroid hormones secreted by the gonads circulate back to the brain to regulate the neural circuits governing the reproductive neuroendocrine axis. These regulatory feedback loops ultimately act to modulate gonadotropin-releasing hormone (GnRH) secretion, thereby affecting gonadotropin secretion from the anterior pituitary. In females, rising estradiol (E2) during the middle of the menstrual (or estrous) cycle paradoxically "switch" from being inhibitory on GnRH secretion ("negative feedback") to stimulating GnRH release ("positive feedback"), resulting in a surge in GnRH secretion and a downstream LH surge that triggers ovulation. While upstream neural afferents of GnRH neurons, including kisspeptin neurons in the rostral hypothalamus, are proposed as critical loci of E2 feedback action, the underlying mechanisms governing the shift between E2 negative and positive feedback are still poorly understood. Indeed, the precise cell targets, neural signaling factors and receptors, hormonal pathways, and molecular mechanisms by which ovarian-derived E2 indirectly stimulates GnRH surge secretion remain incompletely known. In many species, there is also a circadian component to the LH surge, restricting its occurrence to specific times of day, but how the circadian clock interacts with endocrine signals to ultimately time LH surge generation also remains a major gap in knowledge. Here, we focus on classic and recent data from rodent models and discuss the consensus knowledge of the neural players, including kisspeptin, the suprachiasmatic nucleus, and glia, as well as endocrine players, including estradiol and progesterone, in the complex regulation and generation of E2-induced LH surges in females.
Collapse
|
23
|
Jamieson BB, Piet R. Kisspeptin neuron electrophysiology: Intrinsic properties, hormonal modulation, and regulation of homeostatic circuits. Front Neuroendocrinol 2022; 66:101006. [PMID: 35640722 DOI: 10.1016/j.yfrne.2022.101006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 05/05/2022] [Accepted: 05/19/2022] [Indexed: 11/04/2022]
Abstract
The obligatory role of kisspeptin (KISS1) and its receptor (KISS1R) in regulating the hypothalamic-pituitary-gonadal axis, puberty and fertility was uncovered in 2003. In the few years that followed, an impressive body of work undertaken in many species established that neurons producing kisspeptin orchestrate gonadotropin-releasing hormone (GnRH) neuron activity and subsequent GnRH and gonadotropin hormone secretory patterns, through kisspeptin-KISS1R signaling, and mediate many aspects of gonadal steroid hormone feedback regulation of GnRH neurons. Here, we review knowledge accrued over the past decade, mainly in genetically modified mouse models, of the electrophysiological properties of kisspeptin neurons and their regulation by hormonal feedback. We also discuss recent progress in our understanding of the role of these cells within neuronal circuits that control GnRH neuron activity and GnRH secretion, energy balance and, potentially, other homeostatic and reproductive functions.
Collapse
Affiliation(s)
| | - Richard Piet
- Brain Health Research Institute and Department of Biological Sciences, Kent State University, Kent, OH, USA.
| |
Collapse
|
24
|
Sex-specific hypothalamic expression of kisspeptin, gonadotropin releasing hormone, and kisspeptin receptor in progressive demyelination model. J Chem Neuroanat 2022; 123:102120. [PMID: 35718292 DOI: 10.1016/j.jchemneu.2022.102120] [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: 03/25/2022] [Revised: 05/24/2022] [Accepted: 06/13/2022] [Indexed: 11/21/2022]
Abstract
Demyelinating diseases, such as multiple sclerosis, decrease the quality of life of patients and can affect reproduction. Assisted reproductive therapies are available, which although effective, aggravate motor symptoms. For this reason, it is important to determine how the control of the hypothalamus-pituitary-gonadal axis is affected in order to develop better strategies for these patients. One way to determine this is using animal models such as the taiep rat, which shows progressive demyelination of the central nervous system, and was used in the present study to characterize the expression of gonadotrophin-releasing hormone (GnRH), Kisspeptin, and kisspeptin receptor (Kiss1R) and luteinizing hormone (LH) secretion. The expression of kisspeptin, GnRH, and Kiss1R was determined at the hypothalamic level by immunofluorescence and serum LH levels were determined by ELISA. The expression of kisspeptin at the hypothalamic level showed sexual dimorphism, where there was an increase in males and a decrease in females during oestrus. There was no change in the expression of GnRH or kisspeptin receptor, regardless of sex. However, a decrease in serum LH concentration was observed in both sexes. The taiep rat showed changes in the expression of kisspeptin at the hypothalamic level. These changes are different from those reported in the literature with the use of animals with experimental allergic encephalomyelitis, this is because both animal models represent different degrees of progression of multiple sclerosis. Our results suggest that the effects on the hypothalamus-pituitary-gonadal axis depend on the differences between the demyelinating processes, their progression, and even individual factors, and it is thus important that fertility treatments are individualized to maximize therapeutic effects.
Collapse
|
25
|
Goodman RL, Herbison AE, Lehman MN, Navarro VM. Neuroendocrine control of gonadotropin-releasing hormone: Pulsatile and surge modes of secretion. J Neuroendocrinol 2022; 34:e13094. [PMID: 35107859 PMCID: PMC9948945 DOI: 10.1111/jne.13094] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 11/28/2022]
Abstract
The concept that different systems control episodic and surge secretion of gonadotropin-releasing hormone (GnRH) was well established by the time that GnRH was identified and formed the framework for studies of the physiological roles of GnRH, and later kisspeptin. Here, we focus on recent studies identifying the neural mechanisms underlying these two modes of secretion, with an emphasis on their core components. There is now compelling data that kisspeptin neurons in the arcuate nucleus that also contain neurokinin B (NKB) and dynorphin (i.e., KNDy cells) and their projections to GnRH dendrons constitute the GnRH pulse generator in mice and rats. There is also strong evidence for a similar role for KNDy neurons in sheep and goats, and weaker data in monkeys and humans. However, whether KNDy neurons act on GnRH dendrons and/or GnRH soma and dendrites that are found in the mediobasal hypothalamus (MBH) of these species remains unclear. The core components of the GnRH/luteinising hormone surge consist of an endocrine signal that initiates the process and a neural trigger that drives GnRH secretion during the surge. In all spontaneous ovulators, the core endocrine signal is a rise in estradiol secretion from the maturing follicle(s), with the site of estrogen positive feedback being the rostral periventricular kisspeptin neurons in rodents and neurons in the MBH of sheep and primates. There is considerable species variations in the neural trigger, with three major classes. First, in reflex ovulators, this trigger is initiated by coitus and carried to the hypothalamus by neural or vascular pathways. Second, in rodents, there is a time of day signal that originates in the suprachiasmatic nucleus and activates rostral periventricular kisspeptin neurons and GnRH soma and dendrites. Finally, in sheep nitric oxide-producing neurons in the ventromedial nucleus, KNDy neurons and rostral kisspeptin neurons all appear to participate in driving GnRH release during the surge.
Collapse
Affiliation(s)
- Robert L. Goodman
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV, USA
| | - Allan E. Herbison
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Michael N. Lehman
- Brain Health Research Institute, Kent State University, Kent, OH, USA
- Department of Biological Sciences, Kent State University, Kent, OH, USA
| | - Victor M. Navarro
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School and Department of Medicine, Boston, MA, USA
| |
Collapse
|
26
|
McCosh RB, O'Bryne KT, Karsch FJ, Breen KM. Regulation of the gonadotropin-releasing hormone neuron during stress. J Neuroendocrinol 2022; 34:e13098. [PMID: 35128742 PMCID: PMC9232848 DOI: 10.1111/jne.13098] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 01/14/2022] [Accepted: 01/18/2022] [Indexed: 11/30/2022]
Abstract
The effect of stress on reproduction and gonadal function has captivated investigators for almost 100 years. Following the identification of gonadotropin-releasing hormone (GnRH) 50 years ago, a niche research field emerged fixated on how stress impairs this central node controlling downstream pituitary and gonadal function. It is now clear that both episodic GnRH secretion in males and females and surge GnRH secretion in females are inhibited during a variety of stress types. There has been considerable advancement in our understanding of numerous stress-related signaling molecules and their ability to impair reproductive neuroendocrine activity during stress. Recently, much attention has turned to the effects of stress on two populations of kisspeptin neurons: the stimulatory afferents to GnRH neurons that regulate pulsatile and surge-type gonadotropin secretion. Indeed, future work is still required to fully construct the neuroanatomical framework underlying stress effects, directly or indirectly, on GnRH neuron function. The present review evaluates and synthesizes evidence related to stress-related signaling molecules acting directly on GnRH neurons. Here, we review the evidence for and against the action of a handful of signaling molecules as inhibitors of GnRH neuron function, including corticotropin-releasing hormone, urocortins, norepinephrine, cortisol/corticosterone, calcitonin gene-related peptide and arginine-phenylalanine-amide-related peptide-3.
Collapse
Affiliation(s)
- Richard B McCosh
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, La Jolla, CA, USA
| | - Kevin T O'Bryne
- Department of Women and Children's Health, Faculty of Life Sciences and Medicine, King's College London, Guy's Campus, London, UK
| | - Fred J Karsch
- Reproductive Sciences Program and Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Kellie M Breen
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, La Jolla, CA, USA
| |
Collapse
|
27
|
Khan HL, Bhatti S, Sehole Z, Younas H, Nathaniel S, Abbas S, Kaloglu C, Ziders R, Yildiz A, Isa AM. Putative Role of the Kisspeptin/Kiss1R System in Promoting Hypothalamic GnRH Release, Pubertal Maturation, and Regulation of Ovulation Considering the Central Reproductive Axis. FERTILITY & REPRODUCTION 2022. [DOI: 10.1142/s2661318222500062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Kisspeptin is a class of neuropeptides that are the product of the Kiss1 gene. These neuropeptides play an important role in maintaining gonadotropin-releasing hormone (GnRH) levels and their release through hypothalamic neurons. Subsequently, they also play an important role in maintaining gonadotropin levels, as GnRH levels stimulate the release of follicle-stimulating hormone (FSH) and luteinizing hormone (LH), which allow induction of gametogenesis of pubertal maturation. The importance of the Kiss1 gene in reproduction became evident when natural mutations in this gene were discovered, which were associated with hypothalamic hypogonadism (HH) and delayed puberty. Kisspeptin and its KISS1R receptors are expressed in the mammalian ovary. The putative role of the Kisspeptin system in the ovary directly controls oocyte maturation, follicular development, and ovulation in an autocrine and paracrine fashion. These essential facts of kisspeptin and its receptor are necessary to maintain the central reproductive axis.
Collapse
Affiliation(s)
- Haroon Latif Khan
- Lahore Institute of Fertility and Endocrinology, Hameed Latif Hospital, Lahore, Pakistan
| | - Shahzad Bhatti
- Lahore Institute of Fertility and Endocrinology, Hameed Latif Hospital, Lahore, Pakistan
| | - Zirva Sehole
- Department of Biochemistry, Kinnaird College for Women, Lahore, Pakistan
| | - Hooria Younas
- Department of Biochemistry, Kinnaird College for Women, Lahore, Pakistan
| | - Sammar Nathaniel
- Department of Biochemistry, Kinnaird College for Women, Lahore, Pakistan
| | - Sana Abbas
- Lahore Institute of Fertility and Endocrinology, Hameed Latif Hospital, Lahore, Pakistan
| | - Celal Kaloglu
- Department of Histology and Embryology, Cumhuriyet University Faculty of Medicine, Sivas, Turkey
| | - Rachel Ziders
- You Family Fertility Buffalo-Niagara Falls Area, University at Buffalo, Buffalo, NY, USA
| | - Aysegul Yildiz
- Department of Molecular Biology and Genetics, Faculty of Science, Mugla Sitki Kocman University, Kotekli, Mugla, Turkey
| | - Ahmed M. Isa
- Assisted Conception Unit, Obstetrics and Gynecology Department, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
28
|
Abstract
The role of central estrogen in cognitive, metabolic, and reproductive health has long fascinated the lay public and scientists alike. In the last two decades, insight into estrogen signaling in the brain and its impact on female physiology is beginning to catch up with the vast information already established for its actions on peripheral tissues. Using newer methods to manipulate estrogen signaling in hormone-sensitive brain regions, neuroscientists are now identifying the molecular pathways and neuronal subtypes required for controlling sex-dependent energy allocation. However, the immense cellular complexity of these hormone-sensitive brain regions makes it clear that more research is needed to fully appreciate how estrogen modulates neural circuits to regulate physiological and behavioral end points. Such insight is essential for understanding how natural or drug-induced hormone fluctuations across lifespan affect women's health.
Collapse
Affiliation(s)
- Holly A Ingraham
- Department of Cellular and Molecular Pharmacology, School of Medicine, Mission Bay, University of California, San Francisco, California, USA;
| | - Candice B Herber
- Department of Cellular and Molecular Pharmacology, School of Medicine, Mission Bay, University of California, San Francisco, California, USA;
| | - William C Krause
- Department of Cellular and Molecular Pharmacology, School of Medicine, Mission Bay, University of California, San Francisco, California, USA;
| |
Collapse
|
29
|
Sivalingam M, Ogawa S, Trudeau VL, Parhar IS. Conserved functions of hypothalamic kisspeptin in vertebrates. Gen Comp Endocrinol 2022; 317:113973. [PMID: 34971635 DOI: 10.1016/j.ygcen.2021.113973] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 12/23/2021] [Accepted: 12/24/2021] [Indexed: 12/12/2022]
Abstract
Hypothalamic kisspeptin encoded by KISS1/Kiss1 gene emerged as a regulator of the reproductive axis in mammals following the discovery of the kisspeptin receptor (Kissr) and its role in reproduction. Kisspeptin-Kissr systems have been investigated in various vertebrates, and a conserved sequence of kisspeptin-Kissr has been identified in most vertebrate species except in the avian linage. In addition, multiple paralogs of kisspeptin sequences have been identified in the non-mammalian vertebrates. The allegedly conserved role of kisspeptin-Kissr in reproduction became debatable when kiss/kissr genes-deficient zebrafish and medaka showed no apparent effect on the onset of puberty, sexual development, maturation and reproductive capacity. Therefore, it is questionable whether the role of kisspeptin in reproduction is conserved among vertebrate species. Here we discuss from a comparative and evolutional aspect the diverse functions of kisspeptin and its receptor in vertebrates. Primarily this review focuses on the role of hypothalamic kisspeptin in reproductive and non-reproductive functions that are conserved in vertebrate species.
Collapse
Affiliation(s)
- Mageswary Sivalingam
- Brain Research Institute, Jeffery Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Petaling Jaya, Selangor, Malaysia
| | - Satoshi Ogawa
- Brain Research Institute, Jeffery Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Petaling Jaya, Selangor, Malaysia
| | - Vance L Trudeau
- Department of Biology, University of Ottawa, Ottawa, ON, Canada
| | - Ishwar S Parhar
- Brain Research Institute, Jeffery Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Petaling Jaya, Selangor, Malaysia.
| |
Collapse
|
30
|
Shen X, Liu Y, Li XF, Long H, Wang L, Lyu Q, Kuang Y, O’Byrne KT. Optogenetic stimulation of Kiss1 ARC terminals in the AVPV induces surge-like luteinizing hormone secretion via glutamate release in mice. Front Endocrinol (Lausanne) 2022; 13:1036235. [PMID: 36425470 PMCID: PMC9678915 DOI: 10.3389/fendo.2022.1036235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 10/17/2022] [Indexed: 11/10/2022] Open
Abstract
Kisspeptin neurons are mainly located in the arcuate (Kiss1ARC, vis-à-vis the GnRH pulse generator) and anteroventral periventricular nucleus (Kiss1AVPV, vis-à-vis the GnRH surge generator). Kiss1ARC send fibre projections that connect with Kiss1AVPV somata. However, studies focused on the role of Kiss1ARC neurons in the LH surge are limited, and the role of Kiss1ARC projections to AVPV (Kiss1ARC→AVPV) in the preovulatory LH surge is still unknown. To investigate its function, this study used optogenetics to selectively stimulate Kiss1ARC→AVPV and measured changes in circulating LH levels. Kiss1ARC in Kiss-Cre-tdTomato mice were virally infected to express channelrhodopsin-2 proteins, and optical stimulation was applied selectively via a fibre optic cannula in the AVPV. Sustained 20 Hz optical stimulation of Kiss1ARC→AVPV from 15:30 to 16:30 h on proestrus effectively induced an immediate increase in LH reaching peak surge-like levels of around 8 ng/ml within 10 min, followed by a gradual decline to baseline over about 40 min. Stimulation at 10 Hz resulted in a non-significant increase in LH levels and 5 Hz stimulation had no effect in proestrous animals. The 20 Hz stimulation induced significantly higher circulating LH levels on proestrus compared with diestrus or estrus, which suggested that the effect of terminal stimulation is modulated by the sex steroid milieu. Additionally, intra-AVPV infusion of glutamate antagonists, AP5+CNQX, completely blocked the increase on LH levels induced by Kiss1ARC→AVPV terminal photostimulation in proestrous animals. These results demonstrate for the first time that optical stimulation of Kiss1ARC→AVPV induces an LH surge-like secretion via glutamatergic mechanisms. In conclusion, Kiss1ARC may participate in LH surge generation by glutamate release from terminal projections in the AVPV.
Collapse
Affiliation(s)
- Xi Shen
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yali Liu
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiao Feng Li
- Department of Women and Children’s Health, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Hui Long
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Li Wang
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qifeng Lyu
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yanping Kuang
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
- *Correspondence: Kevin T. O’Byrne, ; Yanping Kuang,
| | - Kevin T. O’Byrne
- Department of Women and Children’s Health, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
- *Correspondence: Kevin T. O’Byrne, ; Yanping Kuang,
| |
Collapse
|
31
|
Barabás K, Kovács G, Vértes V, Kövesdi E, Faludi P, Udvarácz I, Pham D, Reglődi D, Abraham IM, Nagy Z. Stereology of gonadotropin-releasing hormone and kisspeptin neurons in PACAP gene-deficient female mice. Front Endocrinol (Lausanne) 2022; 13:993228. [PMID: 36387875 PMCID: PMC9640735 DOI: 10.3389/fendo.2022.993228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 10/05/2022] [Indexed: 11/13/2022] Open
Abstract
The hypothalamic gonadotropin-releasing hormone (GnRH)-kisspeptin neuronal network regulates fertility in all mammals. Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide isolated from the hypothalamus that is involved in the regulation of several releasing hormones and trop hormones. It is well-known that PACAP influences fertility at central and peripheral levels. However, the effects of PACAP on GnRH and kisspeptin neurons are not well understood. The present study investigated the integrity of the estrous cycle in PACAP-knockout (KO) mice. The number and immunoreactivity of GnRH (GnRH-ir) neurons in wild-type (WT) and PACAP KO female mice were determined using immunohistochemistry. In addition, the number of kisspeptin neurons was measured by counting kisspeptin mRNA-positive cells in the rostral periventricular region of the third ventricle (RP3V) and arcuate nucleus (ARC) using the RNAscope technique. Finally, the mRNA and protein expression of estrogen receptor alpha (ERα) was also examined. Our data showed that the number of complete cycles decreased, and the length of each cycle was longer in PACAP KO mice. Furthermore, the PACAP KO mice experienced longer periods of diestrus and spent significantly less time in estrus. There was no difference in GnRH-ir or number of GnRH neurons. In contrast, the number of kisspeptin neurons was decreased in the ARC, but not in the R3PV, in PACAP KO mice compared to WT littermates. Furthermore, ERα mRNA and protein expression was decreased in the ARC, whereas in the R3PV region, ERα mRNA levels were elevated. Our results demonstrate that embryonic deletion of PACAP significantly changes the structure and presumably the function of the GnRH-kisspeptin neuronal network, influencing fertility.
Collapse
Affiliation(s)
- Klaudia Barabás
- Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
- Centre for Neuroscience, Szentágothai Research Centre, Pécs, Hungary
- *Correspondence: Klaudia Barabás, ; Gergely Kovács,
| | - Gergely Kovács
- Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
- Centre for Neuroscience, Szentágothai Research Centre, Pécs, Hungary
- *Correspondence: Klaudia Barabás, ; Gergely Kovács,
| | - Viola Vértes
- Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
| | - Erzsébet Kövesdi
- Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
- Centre for Neuroscience, Szentágothai Research Centre, Pécs, Hungary
| | - Péter Faludi
- Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
- Centre for Neuroscience, Szentágothai Research Centre, Pécs, Hungary
| | - Ildikó Udvarácz
- Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
- Centre for Neuroscience, Szentágothai Research Centre, Pécs, Hungary
| | - Dániel Pham
- Centre for Neuroscience, Szentágothai Research Centre, Pécs, Hungary
- Department of Anatomy, Medical School, University of Pécs, Pécs, Hungary
| | - Dóra Reglődi
- Centre for Neuroscience, Szentágothai Research Centre, Pécs, Hungary
- Department of Anatomy, Medical School, University of Pécs, Pécs, Hungary
| | - Istvan M. Abraham
- Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
- Centre for Neuroscience, Szentágothai Research Centre, Pécs, Hungary
| | - Zsuzsanna Nagy
- Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
| |
Collapse
|
32
|
GnRH neurons recruit astrocytes in infancy to facilitate network integration and sexual maturation. Nat Neurosci 2021; 24:1660-1672. [PMID: 34795451 DOI: 10.1038/s41593-021-00960-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 10/08/2021] [Indexed: 12/25/2022]
Abstract
Neurons that produce gonadotropin-releasing hormone (GnRH), which control fertility, complete their nose-to-brain migration by birth. However, their function depends on integration within a complex neuroglial network during postnatal development. Here, we show that rodent GnRH neurons use a prostaglandin D2 receptor DP1 signaling mechanism during infancy to recruit newborn astrocytes that 'escort' them into adulthood, and that the impairment of postnatal hypothalamic gliogenesis markedly alters sexual maturation by preventing this recruitment, a process mimicked by the endocrine disruptor bisphenol A. Inhibition of DP1 signaling in the infantile preoptic region, where GnRH cell bodies reside, disrupts the correct wiring and firing of GnRH neurons, alters minipuberty or the first activation of the hypothalamic-pituitary-gonadal axis during infancy, and delays the timely acquisition of reproductive capacity. These findings uncover a previously unknown neuron-to-neural-progenitor communication pathway and demonstrate that postnatal astrogenesis is a basic component of a complex set of mechanisms used by the neuroendocrine brain to control sexual maturation.
Collapse
|
33
|
Phumsatitpong C, Wagenmaker ER, Moenter SM. Neuroendocrine interactions of the stress and reproductive axes. Front Neuroendocrinol 2021; 63:100928. [PMID: 34171353 PMCID: PMC8605987 DOI: 10.1016/j.yfrne.2021.100928] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 06/17/2021] [Accepted: 06/19/2021] [Indexed: 01/27/2023]
Abstract
Reproduction is controlled by a sequential regulation of the hypothalamo-pituitary-gonadal (HPG) axis. The HPG axis integrates multiple inputs to maintain proper reproductive functions. It has long been demonstrated that stress alters fertility. Nonetheless, the central mechanisms of how stress interacts with the reproductive system are not fully understood. One of the major pathways that is activated during the stress response is the hypothalamo-pituitary-adrenal (HPA) axis. In this review, we discuss several aspects of the interactions between these two neuroendocrine systems to offer insights to mechanisms of how the HPA and HPG axes interact. We have also included discussions of other systems, for example GABA-producing neurons, where they are informative to the overall picture of stress effects on reproduction.
Collapse
Affiliation(s)
- Chayarndorn Phumsatitpong
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Elizabeth R Wagenmaker
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Suzanne M Moenter
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States; Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, United States.
| |
Collapse
|
34
|
Yeo SH, Herde MK, Herbison AE. Morphological assessment of GABA and glutamate inputs to GnRH neurons in intact female mice using expansion microscopy. J Neuroendocrinol 2021; 33:e13021. [PMID: 34427015 DOI: 10.1111/jne.13021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 01/09/2023]
Abstract
The roles GABAergic and glutamatergic inputs in regulating the activity of the gonadotrophin-releasing hormone (GnRH) neurons at the time of the preovulatory surge remain unclear. We used expansion microscopy to compare the density of GABAergic and glutamatergic synapses on the GnRH neuron cell body and proximal dendrite in dioestrous and pro-oestrous female mice. An evaluation of all synapses immunoreactive for synaptophysin revealed that the highest density of inputs to rostral preoptic area GnRH neurons occurred within the first 45 µm of the primary dendrite (approximately 0.19 synapses µm-1 ) with relatively few synapses on the GnRH neuron soma or beyond 45 µm of the dendrite (0.05-0.08 synapses µm-1 ). Triple immunofluorescence labelling demonstrated a predominance of glutamatergic signalling with twice as many vesicular glutamate transporter 2 synapses detected compared to vesicular GABA transporter. Co-labelling with the GABAA receptor scaffold protein gephyrin and the glutamate receptor postsynaptic density marker Homer1 confirmed these observations, as well as the different spatial distribution of GABA and glutamate inputs along the dendrite. Quantitative assessments revealed no differences in synaptophysin, GABA or glutamate synapses at the proximal dendrite and soma of GnRH neurons between dioestrous and pro-oestrous mice. Taken together, these studies demonstrate that the GnRH neuron receives twice as many glutamatergic synapses compared to GABAergic synapses and that these inputs preferentially target the first 45 µm of the GnRH neuron proximal dendrite. These inputs appear to be structurally stable before the onset of pro-oestrous GnRH surge.
Collapse
Affiliation(s)
- Shel-Hwa Yeo
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Michel K Herde
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Allan E Herbison
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, UK
| |
Collapse
|
35
|
Nandankar N, Negrón AL, Wolfe A, Levine JE, Radovick S. Deficiency of arcuate nucleus kisspeptin results in postpubertal central hypogonadism. Am J Physiol Endocrinol Metab 2021; 321:E264-E280. [PMID: 34181485 PMCID: PMC8410100 DOI: 10.1152/ajpendo.00088.2021] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 06/07/2021] [Accepted: 06/19/2021] [Indexed: 11/25/2022]
Abstract
Kisspeptin (encoded by Kiss1), a neuropeptide critically involved in neuroendocrine regulation of reproduction, is primarily synthesized in two hypothalamic nuclei: the anteroventral periventricular nucleus (AVPV) and arcuate nucleus (ARC). AVPV kisspeptin is thought to regulate the estrogen-induced positive feedback control of gonadotropin-releasing hormone (GnRH) and luteinizing hormone (LH), and the preovulatory LH surge in females. In contrast, ARC kisspeptin neurons, which largely coexpress neurokinin B and dynorphin A (collectively named KNDy neurons), are thought to mediate estrogen-induced negative feedback control of GnRH/LH and be the major regulators of pulsatile GnRH/LH release. However, definitive data to delineate the specific roles of AVPV versus ARC kisspeptin neurons in the control of GnRH/LH release is lacking. Therefore, we generated a novel mouse model targeting deletion of Kiss1 to the ARC nucleus (Pdyn-Cre/Kiss1fl/fl KO) to determine the functional differences between ARC and AVPV kisspeptin neurons on the reproductive axis. The efficacy of the knockout was confirmed at both the mRNA and protein levels. Adult female Pdyn-Cre/Kiss1fl/fl KO mice exhibited persistent diestrus and significantly fewer LH pulses when compared with controls, resulting in arrested folliculogenesis, hypogonadism, and infertility. Pdyn-Cre/Kiss1fl/fl KO males also exhibited disrupted LH pulsatility, hypogonadism, and variable, defective spermatogenesis, and subfertility. The timing of pubertal onset in males and females was equivalent to controls. These findings add to the current body of evidence for the critical role of kisspeptin in ARC KNDy neurons in GnRH/LH pulsatility in both sexes, while directly establishing ARC kisspeptin's role in regulating estrous cyclicity in female mice, and gametogenesis in both sexes, and culminating in disrupted fertility. The Pdyn-Cre/Kiss1fl/fl KO mice present a novel mammalian model of postpubertal central hypogonadism.NEW & NOTEWORTHY We demonstrate through a novel, conditional knockout mouse model of arcuate nucleus (ARC)-specific kisspeptin in the KNDy neuron that ARC kisspeptin is critical for estrous cyclicity in female mice and GnRH/LH pulsatility in both sexes. Our study reveals that ARC kisspeptin is essential for normal gametogenesis, and the loss of ARC kisspeptin results in significant hypogonadism, impacting fertility status. Our findings further confirm that normal puberty occurs despite a loss of ARC kisspeptin.
Collapse
Affiliation(s)
- Nimisha Nandankar
- Department of Pediatrics, Child Health Institute of New Jersey, Rutgers-Robert Wood Johnson Medical School, Rutgers, the State University of New Jersey, New Brunswick, New Jersey
| | - Ariel L Negrón
- Department of Pediatrics, Child Health Institute of New Jersey, Rutgers-Robert Wood Johnson Medical School, Rutgers, the State University of New Jersey, New Brunswick, New Jersey
| | - Andrew Wolfe
- Division of Physiological and Pathological Sciences, National Institutes of Health, Bethesda, Maryland
| | - Jon E Levine
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin
| | - Sally Radovick
- Department of Pediatrics, Child Health Institute of New Jersey, Rutgers-Robert Wood Johnson Medical School, Rutgers, the State University of New Jersey, New Brunswick, New Jersey
| |
Collapse
|
36
|
Delli V, Silva MSB, Prévot V, Chachlaki K. The KiNG of reproduction: Kisspeptin/ nNOS interactions shaping hypothalamic GnRH release. Mol Cell Endocrinol 2021; 532:111302. [PMID: 33964320 DOI: 10.1016/j.mce.2021.111302] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 04/20/2021] [Accepted: 04/23/2021] [Indexed: 12/21/2022]
Abstract
Gonadotropin-releasing hormone (GnRH) is the master regulator of the hypothalamic-pituitary-gonadal (HPG) axis, and therefore of fertility and reproduction. The release pattern of GnRH by the hypothalamus includes both pulses and surges. However, despite a considerable body of evidence in support of a determinant role for kisspeptin, the mechanisms regulating a GnRH pulse and surge remain a topic of debate. In this review we challenge the view of kisspeptin as an absolute "monarch", and instead present the idea of a Kisspeptin-nNOS-GnRH or "KiNG" network that is responsible for generating the "GnRH pulse" and "GnRH surge". In particular, the neuromodulator nitric oxide (NO) has opposite effects to kisspeptin on GnRH secretion in many respects, acting as the Yin to kisspeptin's Yang and creating a dynamic system in which kisspeptin provides the "ON" signal, promoting GnRH release, while NO mediates the "OFF" signal, acting as a tonic brake on GnRH secretion. This interplay between an activator and an inhibitor, which is in turn fine-tuned by the gonadal steroid environment, thus leads to the generation of GnRH pulses and surges and is crucial for the proper development and function of the reproductive axis.
Collapse
Affiliation(s)
- Virginia Delli
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S 1172, F-59000, Lille, France; FHU, 1000 Days for Health, F-59000, Lille, France
| | - Mauro S B Silva
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S 1172, F-59000, Lille, France; FHU, 1000 Days for Health, F-59000, Lille, France
| | - Vincent Prévot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S 1172, F-59000, Lille, France; FHU, 1000 Days for Health, F-59000, Lille, France
| | - Konstantina Chachlaki
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S 1172, F-59000, Lille, France; FHU, 1000 Days for Health, F-59000, Lille, France; University Research Institute of Child Health and Precision Medicine, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece.
| |
Collapse
|
37
|
Gonadotropin-Releasing Hormone (GnRH) Neuron Potassium Currents and Excitability in Both Sexes Exhibit Minimal Changes upon Removal of Negative Feedback. eNeuro 2021; 8:ENEURO.0126-21.2021. [PMID: 34135001 PMCID: PMC8266219 DOI: 10.1523/eneuro.0126-21.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/24/2021] [Accepted: 06/06/2021] [Indexed: 11/24/2022] Open
Abstract
Gonadotropin-releasing hormone (GnRH) drives pituitary secretion of luteinizing hormone and follicle-stimulating hormone, which in turn regulate gonadal functions including steroidogenesis. The pattern of GnRH release and thus fertility depend on gonadal steroid feedback. Under homeostatic (negative) feedback conditions, removal of the gonads from either females or males increases the amplitude and frequency of GnRH release and alters the long-term firing pattern of these neurons in brain slices. The neurobiological mechanisms intrinsic to GnRH neurons that are altered by homeostatic feedback are not well studied and have not been compared between sexes. During estradiol-positive feedback, which is unique to females, there are correlated changes in voltage-gated potassium currents and neuronal excitability. We thus hypothesized that these same mechanisms would be engaged in homeostatic negative feedback. Voltage-gated potassium channels play a direct role in setting excitability and action potential properties. Whole-cell patch-clamp recordings of GFP-identified GnRH neurons in brain slices from sham-operated and castrated adult female and male mice were made to assess fast and slow inactivating potassium currents as well as action potential properties. Surprisingly, no changes were observed among groups in most potassium current properties, input resistance, or capacitance, and this was reflected in a lack of differences in excitability and specific action potential properties. These results support the concept that, in contrast to positive feedback, steroid-negative feedback regulation of GnRH neurons in both sexes is likely conveyed to GnRH neurons via mechanisms that do not induce major changes in the biophysical properties of these cells.
Collapse
|
38
|
Jamieson BB, Bouwer GT, Campbell RE, Piet R. Estrous Cycle Plasticity in the Central Clock Output to Kisspeptin Neurons: Implications for the Preovulatory Surge. Endocrinology 2021; 162:6213415. [PMID: 33824970 DOI: 10.1210/endocr/bqab071] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Indexed: 11/19/2022]
Abstract
Coordination of ovulation and behavior is critical to reproductive success in many species. During the female estrous cycle, the preovulatory gonadotropin surge occurs when ovarian follicles reach maturity and, in rodents, it begins just before the daily onset of activity, ensuring that ovulation coincides with sex behavior. Timing of the surge relies on projections from the suprachiasmatic nucleus (SCN), the locus of the central circadian clock, to hypothalamic circuits that regulate gonadotropin secretion. The cellular mechanisms through which the SCN controls these circuits and gates the preovulatory surge to the appropriate estrous cycle stage, however, are poorly understood. We investigated in mice the functional impact of SCN arginine-vasopressin (AVP) neuron projections to kisspeptin (Kiss1) neurons in the rostral periventricular area of the third ventricle (RP3VKiss1), responsible for generating the preovulatory surge. Conditional anterograde tracing revealed that SCNAVP neurons innervate approximately half of the RP3VKiss1 neurons. Optogenetic activation of SCNAVP projections in brain slices caused an AVP-mediated stimulation of RP3VKiss1 action potential firing in proestrus, the cycle stage when the surge is generated. This effect was less prominent in diestrus, the preceding cycle stage, and absent in estrus, following ovulation. Remarkably, in estrus, activation of SCNAVP projections resulted in GABA-mediated inhibition of RP3VKiss1 neuron firing, an effect rarely encountered in other cycle stages. Together, these data reveal functional plasticity in SCNAVP neuron output that drives opposing effects on RP3VKiss1 neuron activity across the ovulatory cycle. This might contribute to gating activation of the preovulatory surge to the appropriate estrous cycle stage.
Collapse
Affiliation(s)
- Bradley B Jamieson
- Centre for Neuroendocrinology & Department of Physiology, University of Otago, Dunedin 9054, New Zealand
| | - Gregory T Bouwer
- Centre for Neuroendocrinology & Department of Physiology, University of Otago, Dunedin 9054, New Zealand
| | - Rebecca E Campbell
- Centre for Neuroendocrinology & Department of Physiology, University of Otago, Dunedin 9054, New Zealand
| | - Richard Piet
- Centre for Neuroendocrinology & Department of Physiology, University of Otago, Dunedin 9054, New Zealand
- Brain Health Research Institute & Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
| |
Collapse
|
39
|
Gore AC, Thompson LM, Bell M, Mennigen JA. Transgenerational effects of polychlorinated biphenyls: 2. Hypothalamic gene expression in rats†. Biol Reprod 2021; 105:690-704. [PMID: 33824955 DOI: 10.1093/biolre/ioab066] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 03/22/2021] [Accepted: 04/01/2021] [Indexed: 01/07/2023] Open
Abstract
Polychlorinated biphenyls (PCBs) are endocrine-disrupting chemicals (EDCs) with well-established effects on reproduction and behavior in developmentally-exposed (F1) individuals. Because of evidence for transgenerational effects of EDCs on the neuroendocrine control of reproductive physiology, we tested the hypothesis that prenatal PCB exposure leads to unique hypothalamic gene-expression profiles in three generations. Pregnant Sprague-Dawley rats were treated on gestational days 16 and 18 with the PCB mixture Aroclor 1221 (A1221), vehicle (3% DMSO in sesame oil), or estradiol benzoate (EB, 50 μg/kg), the latter a positive control for estrogenic effects of A1221. Maternal- and paternal-lineage F2 and F3 generations were bred using untreated partners. The anteroventral periventricular nucleus (AVPV) and arcuate nucleus (ARC), involved in the hypothalamic control of reproduction, were dissected from F1 to F3 females and males, RNA extracted, and gene expression measured in a qPCR array. We detected unique gene-expression profiles in each generation, which were sex- and lineage-specific. In the AVPV, treatment significantly changed 10, 25, and 11 transcripts in F1, F2, and F3 generations, whereas 10, 1, and 12 transcripts were changed in these generations in the ARC. In the F1 AVPV and ARC, most affected transcripts were decreased by A1221. In the F2 AVPV, most effects of A1221 were observed in females of the maternal lineage, whereas only Pomc expression changed in the F2 ARC (by EB). The F3 AVPV and ARC were mainly affected by EB. It is notable that results in one generation do not predict results in another, and that lineage was a major determinant in results. Thus, transient prenatal exposure of F1 rats to A1221 or EB can alter hypothalamic gene expression across three generations in a sex- and lineage-dependent manner, leading to the conclusion that the legacy of PCBs continues for generations.
Collapse
Affiliation(s)
- Andrea C Gore
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX, USA
| | - Lindsay M Thompson
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX, USA
| | - Mandee Bell
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX, USA
| | - Jan A Mennigen
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
40
|
Owen CM, Zhou X, Bernard DJ, Jaffe LA. Kisspeptin-54 injection induces a physiological luteinizing hormone surge and ovulation in mice. Biol Reprod 2021; 104:1181-1183. [PMID: 33824962 PMCID: PMC8181963 DOI: 10.1093/biolre/ioab067] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/08/2021] [Accepted: 01/12/2021] [Indexed: 01/19/2023] Open
Abstract
Intraperitoneal injection of kisspeptin-54 induces a surge-like release of luteinizing hormone that stimulates ovulation in mice.
Collapse
Affiliation(s)
- Corie M Owen
- Correspondence: Department of Cell Biology, University of Connecticut Health Center, 263 Farmington Ave., MC3636, Farmington, CT 06030 USA. Tel: +1 8606793476; Fax: 860-679-1269
| | - Xiang Zhou
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Canada,Department of Anatomy and Cell Biology, McGill University, Montréal, Canada
| | - Daniel J Bernard
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Canada,Department of Anatomy and Cell Biology, McGill University, Montréal, Canada
| | - Laurinda A Jaffe
- Department of Cell Biology, University of Connecticut Health Center, Farmington CT, USA
| |
Collapse
|
41
|
Zang S, Yin X, Li P. Downregulation of TTF1 in the rat hypothalamic ARC or AVPV nucleus inhibits Kiss1 and GnRH expression, leading to puberty delay. Reprod Biol Endocrinol 2021; 19:30. [PMID: 33622350 PMCID: PMC7901190 DOI: 10.1186/s12958-021-00710-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 02/12/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND TTF1 is a transcription factor that is expressed in the hypothalamus after birth and plays crucial roles in pubertal development. TTF1 may regulate the expression of the Kiss1 gene, which may drive puberty onset in the hypothalamic arcuate (ARC) and anterior ventral paraventricular (AVPV) nuclei. METHODS A dual-luciferase reporter assay was used to detect binding between TTF1 and the Kiss1 gene promoter. To investigate the effects of TTF1, we modified TTF1 expression in cell lines and in the ARC or AVPV nucleus of 21-day-old female rats via lentivirus infection. TTF1 and other puberty onset-related genes were detected by qRT-PCR and western blot analyses. RESULTS The in vitro data indicated that TTF1 knockdown (KD) significantly reduced Kiss1 and GnRH expression. Overexpression (OE) of TTF1 promoted Kiss1 expression. In vivo, the expression of Kiss1 and GnRH decreased significantly in the rats with hypothalamic ARC- or AVPV-specific TTF1 KD. The TTF1-KD rats showed vaginal opening delay. H&E staining revealed that the corpus luteum was obviously reduced at the early puberty and adult stages in the rats with ARC- or AVPV-specific TTF1 KD. CONCLUSION TTF1 bound to the promoter of the Kiss1 gene and enhanced its expression. For 21-day-old female rats, decreased TTF1 in the hypothalamic ARC or AVPV nucleus resulted in delayed vaginal opening and ovarian abnormalities. These observations suggested that TTF1 regulates puberty onset by promoting the expression of Kiss1 and plays an important role in gonad development.
Collapse
Affiliation(s)
- Shaolian Zang
- Department of Endocrinology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, 200062, People's Republic of China
| | - Xiaoqin Yin
- Department of Endocrinology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, 200062, People's Republic of China
| | - Pin Li
- Department of Endocrinology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, 200062, People's Republic of China.
| |
Collapse
|
42
|
McIlwraith EK, Belsham DD. Hypothalamic reproductive neurons communicate through signal transduction to control reproduction. Mol Cell Endocrinol 2020; 518:110971. [PMID: 32750397 DOI: 10.1016/j.mce.2020.110971] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/11/2020] [Accepted: 07/28/2020] [Indexed: 12/11/2022]
Abstract
Gonadotropin-releasing hormone (GnRH) neurons in the hypothalamus coordinate fertility and puberty. In order to achieve successful reproductive capacity, they receive signals from the periphery and from other hypothalamic neurons that coordinate energy homeostasis. Hormones, such as estradiol, insulin, leptin, and adiponectin, act directly or indirectly on GnRH and its associated reproductive neurons. Nutrients like glucose and fatty acids can also affect reproductive neurons to signal nutrient availability. Additionally, acute and chronic inflammation is reported to detrimentally affect GnRH and kisspeptin expression. All of these cues activate signal transduction pathways within neurons that lead to the changes in GnRH neuronal function. The signalling pathways can also be dysregulated by endocrine disrupting chemicals, which impair fertility by misappropriating common signalling pathways. The complex mechanisms controlling the levels of GnRH during the reproductive cycle rely on a carefully orchestrated set of signal transduction events to regulate the positive and negative feedback arms of the hypothalamic-pituitary-gonadal axis. If these signalling events are dysregulated, this will result is a downregulatory event leading to hypogonadal hypogonadism with decreased or absent fertility. Therefore, an understanding of the mechanisms involved in distinct neuronal signalling could provide an advantage to inform therapeutic interventions for infertility and reproductive disorders.
Collapse
Affiliation(s)
- Emma K McIlwraith
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Denise D Belsham
- Department of Physiology, University of Toronto, Toronto, ON, Canada; Obstetrics and Gynaecology and Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
43
|
Phumsatitpong C, De Guzman RM, Zuloaga DG, Moenter SM. A CRH Receptor Type 1 Agonist Increases GABA Transmission to GnRH Neurons in a Circulating-Estradiol-Dependent Manner. Endocrinology 2020; 161:5892962. [PMID: 32798220 PMCID: PMC7547842 DOI: 10.1210/endocr/bqaa140] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 08/06/2020] [Indexed: 12/15/2022]
Abstract
GnRH neurons are central regulators of reproduction and respond to factors affecting fertility, such as stress. Corticotropin-releasing hormone (CRH) is released during stress response. In brain slices from unstressed controls, CRH has opposite, estradiol-dependent effects on GnRH neuron firing depending on the CRH receptor activated; activating CRHR-1 stimulates whereas activating CRHR-2 suppresses activity. We investigated possible direct and indirect mechanisms. Mice were ovariectomized and either not treated further (OVX) or given a capsule producing high positive feedback (OVX + E) or low negative feedback (OVX + low E) physiologic circulating estradiol levels. We tested possible direct effects on GnRH neurons by altering voltage-gated potassium currents. Two types of voltage-gated potassium currents (transient IA and sustained IK) were measured; neither CRHR-1 nor CRHR-2 agonists altered potassium current density in GnRH neurons from OVX + E mice. Further, neither CRH nor receptor-specific agonists altered action potential generation in response to current injection in GnRH neurons from OVX + E mice. To test the possible indirect actions, GABAergic postsynaptic currents were monitored. A CRHR-1 agonist increased GABAergic transmission frequency to GnRH neurons from OVX + E, but not OVX, mice, whereas a CRHR-2 agonist had no effect. Finally, we tested if CRH alters the firing rate of arcuate kisspeptin neurons, which provide an important excitatory neuromodulatory input to GnRH neurons. CRH did not acutely alter firing activity of these neurons from OVX, OVX + E or OVX + low E mice. These results suggest CRH increases GnRH neuron activity in an estradiol-dependent manner in part by activating GABAergic afferents. Mechanisms underlying inhibitory effects of CRH remain unknown.
Collapse
Affiliation(s)
| | | | | | - Suzanne M Moenter
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, US
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, US
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, US
- Correspondence: Suzanne M. Moenter; 7725 Med Sci II; 1137 E Catherine St; Ann Arbor, MI 48109-5622. E-mail:
| |
Collapse
|
44
|
Liu Y, Li X, Shen X, Ivanova D, Lass G, He W, Chen Q, Yu S, Wang Y, Long H, Wang L, Lyu Q, Kuang Y, O’Byrne KT. Dynorphin and GABAA Receptor Signaling Contribute to Progesterone's Inhibition of the LH Surge in Female Mice. Endocrinology 2020; 161:5808894. [PMID: 32181477 PMCID: PMC7153819 DOI: 10.1210/endocr/bqaa036] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 02/28/2020] [Indexed: 01/02/2023]
Abstract
Progesterone can block estrogen-induced luteinising hormone (LH) surge secretion and can be used clinically to prevent premature LH surges. The blocking effect of progesterone on the LH surge is mediated through its receptor in the anteroventral periventricular nucleus (AVPV) of the hypothalamus. However, the underlying mechanisms are unclear. The preovulatory LH surge induced by estrogen is preceded by a significant reduction in hypothalamic dynorphin and gamma-aminobutyric acid (GABA) release. To test the detailed roles of dynorphin and GABA in an LH surge blockade by progesterone, ovariectomized and 17β-estradiol capsule-implanted (OVX/E2) mice received simultaneous injections of estradiol benzoate (EB) and progesterone (P) or vehicle for 2 consecutive days. The LH level was monitored from 2:30 pm to 8:30 pm at 30-minute intervals. Progesterone coadministration resulted in the LH surge blockade. A continuous microinfusion of the dynorphin receptor antagonist nor-BNI or GABAA receptor antagonist bicuculline into the AVPV from 3:00 pm to 7:00 pm reversed the progesterone-mediated blockade of the LH surge in 7 of 9 and 6 of 10 mice, respectively. In addition, these LH surges started much earlier than the surge induced by estrogen alone. However, 5 of 7 progesterone-treated mice did not show LH surge secretion after microinfusion with the GABAB receptor antagonist CGP-35348. Additionally, peripheral administration of kisspeptin-54 promotes LH surge-like release in progesterone treated mice. These results demonstrated that the progesterone-mediated suppression of the LH surge is mediated by an increase in dynorphin and GABAA receptor signaling acting though kisspeptin neurons in the AVPV of the hypothalamus in female mice.
Collapse
Affiliation(s)
- Yali Liu
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Huangpu District, Shanghai, China
- Department of Women and Children’s Health, Faculty of Life Sciences and Medicine, King’s College London, Guy’s Campus, UK
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Pudong New Area, Shanghai, China
| | - Xiaofeng Li
- Department of Women and Children’s Health, Faculty of Life Sciences and Medicine, King’s College London, Guy’s Campus, UK
| | - Xi Shen
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Huangpu District, Shanghai, China
- Department of Women and Children’s Health, Faculty of Life Sciences and Medicine, King’s College London, Guy’s Campus, UK
| | - Deyana Ivanova
- Department of Women and Children’s Health, Faculty of Life Sciences and Medicine, King’s College London, Guy’s Campus, UK
| | - Geffen Lass
- Department of Women and Children’s Health, Faculty of Life Sciences and Medicine, King’s College London, Guy’s Campus, UK
| | - Wen He
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Huangpu District, Shanghai, China
| | - Qiuju Chen
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Huangpu District, Shanghai, China
| | - Sha Yu
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Huangpu District, Shanghai, China
| | - Yun Wang
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Huangpu District, Shanghai, China
| | - Hui Long
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Huangpu District, Shanghai, China
| | - Li Wang
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Huangpu District, Shanghai, China
| | - Qifeng Lyu
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Huangpu District, Shanghai, China
| | - Yanping Kuang
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Huangpu District, Shanghai, China
- Correspondence: Kevin O’Byrne, PhD, 2.92W Hodgkin Building, Department of Women and Children’s Health, Faculty of Life Sciences and Medicine, Guy’s Campus, King’s College London, London, SE1 1UL. E-mail: ; or Yanping Kuang, Department of Assisted Reproduction, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People’s Republic of China. E-mail:
| | - Kevin T O’Byrne
- Department of Women and Children’s Health, Faculty of Life Sciences and Medicine, King’s College London, Guy’s Campus, UK
- Correspondence: Kevin O’Byrne, PhD, 2.92W Hodgkin Building, Department of Women and Children’s Health, Faculty of Life Sciences and Medicine, Guy’s Campus, King’s College London, London, SE1 1UL. E-mail: ; or Yanping Kuang, Department of Assisted Reproduction, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People’s Republic of China. E-mail:
| |
Collapse
|
45
|
Herbison AE. A simple model of estrous cycle negative and positive feedback regulation of GnRH secretion. Front Neuroendocrinol 2020; 57:100837. [PMID: 32240664 DOI: 10.1016/j.yfrne.2020.100837] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 03/19/2020] [Accepted: 03/19/2020] [Indexed: 12/19/2022]
Abstract
The gonadal steroids estradiol and progesterone exert critical suppressive and stimulatory actions upon the brain to control gonadotropin-releasing hormone (GnRH) release that drives the estrous/menstrual cycle. A simple model for understanding these interactions is proposed in which the activity of the "GnRH pulse generator" is restrained by post-ovulation progesterone secretion to bring about the estrus/luteal phase slowing of pulsatile gonadotropin release, while the activity of the "GnRH surge generator" is primed by the rising follicular phase levels of estradiol to generate the pre-ovulatory surge. The physiological fluctuations in estradiol levels across the cycle are considered to clamp the GnRH pulse generator output at a constant level. Independent pulse and surge generator circuitries regulate the excitability of different compartments of the GnRH neuron. As such, GnRH secretion through the cycle is determined simply by the summed influence of the estradiol-clamped, progesterone-regulated pulse and estradiol-regulated surge generators on the GnRH neuron.
Collapse
Affiliation(s)
- Allan E Herbison
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin 9054, New Zealand.
| |
Collapse
|
46
|
Sen A, Hoffmann HM. Role of core circadian clock genes in hormone release and target tissue sensitivity in the reproductive axis. Mol Cell Endocrinol 2020; 501:110655. [PMID: 31756424 PMCID: PMC6962569 DOI: 10.1016/j.mce.2019.110655] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 11/15/2019] [Accepted: 11/16/2019] [Indexed: 12/17/2022]
Abstract
Precise timing in hormone release from the hypothalamus, the pituitary and ovary is critical for fertility. Hormonal release patterns of the reproductive axis are regulated by a feedback loop within the hypothalamic-pituitary-gonadal (HPG) axis. The timing and rhythmicity of hormone release and tissue sensitivity in the HPG axis is regulated by circadian clocks located in the hypothalamus (suprachiasmatic nucleus, kisspeptin and GnRH neurons), the pituitary (gonadotrophs), the ovary (theca and granulosa cells), the testis (Leydig cells), as well as the uterus (endometrium and myometrium). The circadian clocks integrate environmental and physiological signals to produce cell endogenous rhythms generated by a transcriptional-translational feedback loop of transcription factors that are collectively called the "molecular clock". This review specifically focuses on the contribution of molecular clock transcription factors in regulating hormone release patterns in the reproductive axis, with an emphasis on the female reproductive system. Specifically, we discuss the contributions of circadian rhythms in distinct neuronal populations of the female hypothalamus, the molecular clock in the pituitary and its overall impact on female and male fertility.
Collapse
Affiliation(s)
- Aritro Sen
- Department of Animal Science and the Reproductive and Developmental Science Program, Michigan State University, East Lansing, MI, 48824, USA
| | - Hanne M Hoffmann
- Department of Animal Science and the Reproductive and Developmental Science Program, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
47
|
Moenter SM, Silveira MA, Wang L, Adams C. Central aspects of systemic oestradiol negative- and positive-feedback on the reproductive neuroendocrine system. J Neuroendocrinol 2020; 32:e12724. [PMID: 31054210 PMCID: PMC6829026 DOI: 10.1111/jne.12724] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 04/24/2019] [Accepted: 04/26/2019] [Indexed: 12/24/2022]
Abstract
The central nervous system regulates fertility via the release of gonadotrophin-releasing hormone (GnRH). This control revolves around the hypothalamic-pituitary-gonadal axis, which operates under traditional homeostatic feedback by sex steroids from the gonads in males and most of the time in females. An exception is the late follicular phase in females, when homeostatic feedback is suspended and a positive-feedback response to oestradiol initiates the preovulatory surges of GnRH and luteinising hormone. Here, we briefly review the history of how mechanisms underlying central control of ovulation by circulating steroids have been studied, discuss the relative merit of different model systems and integrate some of the more recent findings in this area into an overall picture of how this phenomenon occurs.
Collapse
Affiliation(s)
- Suzanne M. Moenter
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, 48109
| | - Marina A. Silveira
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109
| | - Luhong Wang
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109
| | - Caroline Adams
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109
| |
Collapse
|
48
|
Conde K, Roepke TA. 17β-Estradiol Increases Arcuate KNDy Neuronal Sensitivity to Ghrelin Inhibition of the M-Current in Female Mice. Neuroendocrinology 2020; 110:582-594. [PMID: 31484184 PMCID: PMC7056582 DOI: 10.1159/000503146] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 09/02/2019] [Indexed: 11/19/2022]
Abstract
Obesity and anorexia result in dysregulation of the hypothalamic-pituitary-gonadal axis, negatively impacting reproduction. Ghrelin, secreted from the stomach, potentially mediates negative energy states and neuroendocrine control of reproduction by acting through the growth hormone secretagogue receptor (GHSR). GHSR is expressed in hypothalamic arcuate (ARC) Kisspeptin/Neurokinin B (Tac2)/Dynorphin (KNDy) neurons. Ghrelin is known to inhibit the M-current produced by KCNQ channels in other ARC neurons. In addition, we have shown 17β-estradiol (E2) increases Ghsr expression in KNDy neurons 6-fold and increases the M-current in NPY neurons. We hypothesize that E2 increases GHSR expression in KNDy neurons to increase ghrelin sensitivity during negative energy states. Furthermore, we suspect ghrelin targets the M-current in KNDy neurons to control reproduction and energy homeostasis. We utilized ovariectomized Tac2-EGFP adult female mice, pretreated with estradiol benzoate (EB) or oil vehicle and performed whole-cell-patch-clamp recordings to elicit the M-current in KNDy neurons using standard activation protocols in voltage-clamp. Using the selective KCNQ channel blocker XE-991 (40 µM) to target the M-current, oil- and EB-treated mice showed a decrease in the maximum peak current by 75.7 ± 13.8 pA (n = 10) and 68.0 ± 14.7 pA (n = 11), respectively. To determine the actions of ghrelin on the M-current, ghrelin was perfused (100 nM) in oil- and EB-treated mice resulting in the suppression of the maximum peak current by 58.5 ± 15.8 pA (n = 9) and 59.2 ± 11.9 pA (n = 9), respectively. KNDy neurons appeared more sensitive to ghrelin when pretreated with EB, revealing that ARC KNDy neurons are more sensitive to ghrelin during states of high E2.
Collapse
Affiliation(s)
- Kristie Conde
- Graduate Program in Neuroscience, Rutgers University Robert Wood Johnson Medical School, The State University of New Jersey, New Brunswick, New Jersey, USA
| | - Troy A Roepke
- Graduate Program in Neuroscience, Rutgers University Robert Wood Johnson Medical School, The State University of New Jersey, New Brunswick, New Jersey, USA,
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA,
| |
Collapse
|
49
|
Wang L, Moenter SM. Differential Roles of Hypothalamic AVPV and Arcuate Kisspeptin Neurons in Estradiol Feedback Regulation of Female Reproduction. Neuroendocrinology 2020; 110:172-184. [PMID: 31466075 PMCID: PMC7047625 DOI: 10.1159/000503006] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 08/28/2019] [Indexed: 11/19/2022]
Abstract
Mammalian reproductive function includes puberty onset and completion, reproductive cyclicity, steroidogenesis, gametogenesis, fertilization, pregnancy, and lactation; all are indispensable to perpetuate species. Reproductive cycles are critical for providing the hormonal milieu needed for follicular development and maturation of eggs, but cycles, in and of themselves, do not guarantee ovulation will occur. Here, we review the roles in female reproductive neuroendocrine function of two hypothalamic populations that produce the neuropeptide kisspeptin, demonstrating distinct roles in maintaining cycles and ovulation.
Collapse
Affiliation(s)
- Luhong Wang
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Suzanne M Moenter
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA,
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA,
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA,
| |
Collapse
|
50
|
Neurochemical Characterization of Neurons Expressing Estrogen Receptor β in the Hypothalamic Nuclei of Rats Using in Situ Hybridization and Immunofluorescence. Int J Mol Sci 2019; 21:ijms21010115. [PMID: 31877966 PMCID: PMC6981915 DOI: 10.3390/ijms21010115] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/14/2019] [Accepted: 12/18/2019] [Indexed: 12/11/2022] Open
Abstract
Estrogens play an essential role in multiple physiological functions in the brain, including reproductive neuroendocrine, learning and memory, and anxiety-related behaviors. To determine these estrogen functions, many studies have tried to characterize neurons expressing estrogen receptors known as ERα and ERβ. However, the characteristics of ERβ-expressing neurons in the rat brain still remain poorly understood compared to that of ERα-expressing neurons. The main aim of this study is to determine the neurochemical characteristics of ERβ-expressing neurons in the rat hypothalamus using RNAscope in situ hybridization (ISH) combined with immunofluorescence. Strong Esr2 signals were observed especially in the anteroventral periventricular nucleus (AVPV), bed nucleus of stria terminalis, hypothalamic paraventricular nucleus (PVN), supraoptic nucleus, and medial amygdala, as previously reported. RNAscope ISH with immunofluorescence revealed that more than half of kisspeptin neurons in female AVPV expressed Esr2, whereas few kisspeptin neurons were found to co-express Esr2 in the arcuate nucleus. In the PVN, we observed a high ratio of Esr2 co-expression in arginine-vasopressin neurons and a low ratio in oxytocin and corticotropin-releasing factor neurons. The detailed neurochemical characteristics of ERβ-expressing neurons identified in the current study can be very essential to understand the estrogen signaling via ERβ.
Collapse
|