1
|
Bearss RJ, Oliver IA, Neuman PN, Abdulmajeed WI, Ackerman JM, Piet R. Activation of ionotropic and group I metabotropic glutamate receptors stimulates kisspeptin neuron activity in mice. J Neuroendocrinol 2024:e13456. [PMID: 39414384 DOI: 10.1111/jne.13456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/18/2024]
Abstract
Different populations of hypothalamic kisspeptin (KISS1) neurons located in the rostral periventricular area of the third ventricle (RP3V) and arcuate nucleus (ARC) are thought to generate the sex-specific patterns of gonadotropin secretion. These neuronal populations integrate gonadal sex steroid feedback with internal and external cues relayed via the actions of neurotransmitters and neuropeptides. The excitatory amino acid neurotransmitter glutamate, the main excitatory neurotransmitter in the brain, plays a role in regulating gonadotropin secretion, at least partially through engaging KISS1 signaling. The expression and function of individual glutamate receptor subtypes in KISS1 neurons, however, are not well characterized. Here, we used GCaMP-based calcium imaging and patch-clamp electrophysiology to assess the impact of activating individual ionotropic (iGluR) and group I metabotropic (mGluR) glutamate receptors on KISS1 neuron activity in the mouse RP3V and ARC. Our results indicate that activation of all iGluR subtypes and of group I mGluRs, likely mGluR1, consistently drives activity in the majority of KISS1 neurons within the RP3V and ARC of males and females. Our results also revealed, somewhat unexpectedly, sex- and region-specific differences. Indeed, activating (S)-α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) type iGluRs evoked larger responses in female ARCKISS1 neurons than in their male counterparts whereas activating group I mGluRs induced larger responses in RP3VKISS1 neurons than in ARCKISS1 neurons in females. Together, our findings suggest that glutamatergic neurotransmission in KISS1 neurons, and its impact on the activity of these cells, might be sex- and region-dependent in mice.
Collapse
Affiliation(s)
- Robin J Bearss
- School of Biomedical Sciences, Kent State University, Kent, Ohio, USA
- Brain Health Research Institute, Kent State University, Kent, Ohio, USA
| | - Isabella A Oliver
- Brain Health Research Institute, Kent State University, Kent, Ohio, USA
| | - Peighton N Neuman
- Brain Health Research Institute, Kent State University, Kent, Ohio, USA
- Department of Biological Sciences, Kent State University, Kent, Ohio, USA
| | - Wahab I Abdulmajeed
- Brain Health Research Institute, Kent State University, Kent, Ohio, USA
- Department of Biological Sciences, Kent State University, Kent, Ohio, USA
| | - Jennifer M Ackerman
- School of Biomedical Sciences, Kent State University, Kent, Ohio, USA
- Brain Health Research Institute, Kent State University, Kent, Ohio, USA
| | - Richard Piet
- School of Biomedical Sciences, Kent State University, Kent, Ohio, USA
- Brain Health Research Institute, Kent State University, Kent, Ohio, USA
- Department of Biological Sciences, Kent State University, Kent, Ohio, USA
| |
Collapse
|
2
|
Villa PA, Ruggiero-Ruff RE, Jamieson BB, Campbell RE, Coss D. Obesity Alters POMC and Kisspeptin Neuron Cross Talk Leading to Reduced Luteinizing Hormone in Male Mice. J Neurosci 2024; 44:e0222242024. [PMID: 38744532 PMCID: PMC11236585 DOI: 10.1523/jneurosci.0222-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/19/2024] [Accepted: 05/07/2024] [Indexed: 05/16/2024] Open
Abstract
Obesity is associated with hypogonadism in males, characterized by low testosterone and sperm number. Previous studies determined that these stem from dysregulation of hypothalamic circuitry that regulates reproduction, by unknown mechanisms. Herein, we used mice fed chronic high-fat diet, which mimics human obesity, to determine mechanisms of impairment at the level of the hypothalamus, in particular gonadotropin-releasing hormone (GnRH) neurons that regulate luteinizing hormone (LH), which then regulates testosterone. Consistent with obese humans, we demonstrated lower LH, and lower pulse frequency of LH secretion, but unchanged pituitary responsiveness to GnRH. LH pulse frequency is regulated by pulsatile GnRH secretion, which is controlled by kisspeptin. Peripheral and central kisspeptin injections, and DREADD-mediated activation of kisspeptin neurons, demonstrated that kisspeptin neurons were suppressed in obese mice. Thus, we investigated regulators of kisspeptin secretion. We determined that the LH response to NMDA was lower in obese mice, corresponding to fewer glutamate receptors in kisspeptin neurons, which may be critical for kisspeptin synchronization. Given that kisspeptin neurons also interact with anorexigenic POMC neurons, which are affected by obesity, we examined their cross talk, and determined that the LH response to either DREADD-mediated activation of POMC neurons or central injection of αMSH, a product of POMC, is abolished in obese mice. This was accompanied by diminished levels of αMSH receptor, MC4R, in kisspeptin neurons. Together, our studies determined that obesity leads to the downregulation of receptors that regulate kisspeptin neurons, which is associated with lower LH pulse frequency, leading to lower LH and hypogonadism.
Collapse
Affiliation(s)
- Pedro A Villa
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California 92521
| | - Rebecca E Ruggiero-Ruff
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California 92521
| | - Bradley B Jamieson
- Centre for Neuroendocrinology, and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Rebecca E Campbell
- Centre for Neuroendocrinology, and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Djurdjica Coss
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California 92521
| |
Collapse
|
3
|
Абсатарова ЮС, Евсеева ЮС, Андреева ЕН. [Neuroendocrine features of the pathogenesis of polycystic ovary syndrome (literature review)]. PROBLEMY ENDOKRINOLOGII 2023; 69:107-114. [PMID: 37968958 PMCID: PMC10680539 DOI: 10.14341/probl13350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 11/17/2023]
Abstract
Polycystic ovary syndrome (PCOS) is one of the most pressing problems in endocrine gynecology. The main signs of the disease are hyperandrogenism, menstrual and/or ovulatory dysfunction, and polycystic ovarian structure according to ultrasound. Women with PCOS are at risk for developing metabolic syndrome, type 2 diabetes, cardiovascular disease, and endometrial cancer. In this connection, the pathogenetic mechanisms of the occurrence of this syndrome are continuously studied and new methods of treatment are being sought. PCOS is characterized by a wide range of various disorders of the neuroendocrine regulation of the reproductive system. The main focus of the review is aimed at summarizing information about the etiological role of neuropeptides and neurotransmitters, such as phoenixin, galanins, orexins, GABA, in the pathophysiology of PCOS and about the possibility of their use for diagnostic and therapeutic purposes. In recent decades, the interest of scientists has been focused on the study of KNDy neurons, because it is the kisspeptin synthesized by them that is one of the main regulators of the hypothalamic-pituitary-ovarian axis. This article discusses data on the significance of KNDy neurons in the pathogenesis of the syndrome. Information is provided on the effect of elevated levels of androgens and anti-Müllerian hormone on GnRH neurons. Also analyzed are studies on functional and structural disorders in the hypothalamus in PCOS. Literature search was carried out in national (eLibrary, CyberLeninka.ru) and international (PubMed, Cochrane Library) databases in Russian and English. The priority was free access to the full text of articles. The choice of sources was prioritized for the period from 2018 to 2023.However, taking into account the insufficient knowledge of the chosen topic, the choice of sources dates back to 1998.
Collapse
Affiliation(s)
| | - Ю. С. Евсеева
- Национальный медицинский исследовательский центр эндокринологии
| | - Е. Н. Андреева
- Национальный медицинский исследовательский центр эндокринологии; Московский государственный медико-стоматологический университет имени А.И. Евдокимова
| |
Collapse
|
4
|
Louden ED, Dougherty MP, Chorich LP, Eroglu A, Layman LC. Investigation of subfertility in the female Nsmf knockout mouse. F&S SCIENCE 2023; 4:286-293. [PMID: 37516276 DOI: 10.1016/j.xfss.2023.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 07/31/2023]
Abstract
OBJECTIVE To study if a pituitary or ovarian defect contributes to subfertility of the female Nsmf knockout (KO) mouse, an animal model of the hypogonadotropic hypogonadism gene NSMF. DESIGN Analysis of hypothalamic, pituitary and ovarian gene expression at baseline, serum gonadotropin levels before and after gonadotropin-releasing hormone (GnRH) stimulation, ovarian response and implantation after superovulation, gonadotropin effects after ovariectomy, and ovarian NSMF protein expression. SETTING University research laboratory. PATIENTS None; mice were used. INTERVENTIONS Gonadotropin-releasing hormone stimulation, superovulation, and ovariectomy in separate experiments. MAIN OUTCOME MEASURES Gene expression in the hypothalamus, pituitary, and ovary; ovarian response and implantation after superovulation; serum gonadotropins after GnRH stimulation and ovariectomy; Western blot to measure ovarian NSMF expression. RESULTS We found increased hypothalamic Kiss1, Gnrh1, and Jak2 mRNA expression in female Nsmf KO vs. wild type (WT) mice. However, pituitary gonadotropin, and GnRH receptor gene expression was not affected, and serum gonadotropin levels were normal. Gonadotropins increased after ovariectomy for both groups. Baseline Kiss1, Fshr, Prkaca, Prkar1a, and Gdf9 ovarian mRNA expression was increased and Cyp19a1 expression was decreased in Nsmf KO mice, while superovulated Nsmf KO mice had reduced ovarian Kiss1r, Prkar1a, and Fshr mRNA expression, 50% less oocytes, and normal implantation. Western blot demonstrated NSMF protein expression in the ovary of WT mice. CONCLUSIONS Altered hypothalamic and ovarian gene expression was demonstrated in female Nsmf KO mice. It is possible that increased hypothalamic Gnrh1 and Kiss1 mRNA expression could compensate for reduced NSMF enabling a normal pituitary gonadotropin response. Impaired superovulation response, altered ovarian gene expression, and decreased number of oocytes indicate ovarian dysfunction, but a uterine factor cannot be excluded. These findings provide an anatomic basis for future mechanistic studies of subfertility in female Nsmf KO mice.
Collapse
Affiliation(s)
- Erica D Louden
- Section of Reproductive Endocrinology, Infertility, & Genetics, Department of Obstetrics & Gynecology
| | - Michael P Dougherty
- Section of Reproductive Endocrinology, Infertility, & Genetics, Department of Obstetrics & Gynecology
| | - Lynn P Chorich
- Section of Reproductive Endocrinology, Infertility, & Genetics, Department of Obstetrics & Gynecology
| | - Ali Eroglu
- Section of Reproductive Endocrinology, Infertility, & Genetics, Department of Obstetrics & Gynecology; Department of Neuroscience and Regenerative Medicine
| | - Lawrence C Layman
- Section of Reproductive Endocrinology, Infertility, & Genetics, Department of Obstetrics & Gynecology; Department of Neuroscience and Regenerative Medicine; Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia.
| |
Collapse
|
5
|
Giannitsioti E, Koutserimpas C, Samonis G, Papadopoulos A. Foot Osteomyelitis Caused by Multidrug- and Extensively Drug-Resistant Gram-Negative Bacteria. MAEDICA 2023; 18:19-26. [PMID: 37266476 PMCID: PMC10231173 DOI: 10.26574/maedica.2023.18.1.19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
There are scarce data regarding foot osteomyelitis (FO) caused by multidrug-resistant (MDR) and extensively drug-resistant (XDR) Gram-negative bacteria (GNB). This study investigates the causative organisms, the diagnostic and therapeutic approach and the outcome of these infections. All patients with FO caused by MDR and/or XDR GNB who received treatment in the Department of osseous infectious diseases of the "Attikon" University Hospital of Athens, Greece, were recorded and evaluated during a six-year-period. Seventeen patients, of which 64.7% females, with a mean age of 54.06 years were studied. There were nine cases with diabetic (DFO), and 76.5% of patients reported previous use of antimicrobials. Pathogens were isolated from soft-tissue biopsies or intra-operative tissue samples (n=12) and/or affected bone samples (n=5). In most cases, E. coli and P. aeruginosa were isolated (each 29.4%), followed by P. mirabilis (11.7%), while polymicrobial infection was detected in nine patients (53%). Most cases were given antimicrobial monotherapy (88.2%) with a mean duration of 90.05 days, while surgery significantly promoted cure of the infection. Foot and DFO cases represent a challenging to treat disease, requiring a multidisciplinary approach. Surgical treatment remains the cornerstone of treatment, while it is of utmost importance to isolate the causative organisms. MDR and XDR GNB represent an emerging threat and more data are needed to better understand these infections.
Collapse
Affiliation(s)
- Efthymia Giannitsioti
- Osseous Infectious Diseases Department, "Attikon" General University Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece, PC: 124 62
| | - Christos Koutserimpas
- Department of Orthopaedics and Traumatology, "251" Hellenic Air Force General Hospital of Athens, Greece, PC:11525
| | - George Samonis
- Department of Medicine, University of Crete, Heraklion, Greece, PC: 715 00
- First Department of Medical Oncology, "Metropolitan" Hospital, Neon Faliron, Attica, Greece, PC: 185 47
| | - Antonios Papadopoulos
- First Department of Medical Oncology, "Metropolitan" Hospital, Neon Faliron, Attica, Greece, PC: 185 47
| |
Collapse
|
6
|
Leonardi CEP, Carrasco RA, Dias FCF, Zwiefelhofer EM, Adams GP, Singh J. Mechanism of LH release after peripheral administration of kisspeptin in cattle. PLoS One 2022; 17:e0278564. [PMID: 36459509 PMCID: PMC9718405 DOI: 10.1371/journal.pone.0278564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 11/18/2022] [Indexed: 12/04/2022] Open
Abstract
Kisspeptin modulates GnRH secretion in mammals and peripheral administration of 10-amino acid fragment of kisspeptin (Kp10) induces LH release and ovulation in cattle. Experiments were done to determine if iv administration of kisspeptin will activate GnRH neurons (i.e., after crossing the blood-brain barrier) and if pre-treatment with a GnRH receptor blocker will alter kisspeptin-induced LH release (from gonadotrophs) and ovulation. In Experiment 1, cows (n = 3 per group) were given human-Kisspeptin10 (hKp10; 3 x 15 mg iv at 60-min intervals) or normal saline and euthanized 150 min after treatment was initiated. Every 20th free-floating section (50 μm thickness) from the preoptic area to hypothalamus was double immunostained to colocalize GnRH- (DAB) and activated neurons (cFOS; Nickel-DAB). Kisspeptin induced plasma LH release from 15 to 150 min (P = 0.01) but the proportion of activated GnRH neurons did not differ between groups (5.8% and 3.5%, respectively; P = 0.11). Immunogold electron microscopy detected close contacts between kisspeptin fibers and GnRH terminals in the median eminence. In Experiment 2, pubertal heifers (n = 5 per group) were treated with 1) hKp10 iv, 2) Cetrorelix (GnRH antagonist; im) + hKp10 iv or 3) saline on Day 6 of the follicular wave under low-progesterone condition. A rise in plasma LH concentration was detected from 15 to 240 min in the hKp10 group but not in cetrorelix or control group (P<0.001). Ovulations were detected only in the hKp10 group (4/5; P = 0.02). Cetrorelix treatment was associated with regression of the preovulatory dominant follicle and emergence of a new follicular wave 3.4±0.75 days after the treatment in all five heifers. Results support the hypothesis that the effect of peripheral kisspeptin is mediated downstream of GnRH synthesis and does not involve GnRH-independent LH release from gonadotrophs. Peripheral kisspeptin may release pre-synthesized GnRH from the nerve terminals in areas outside the blood-brain barrier.
Collapse
Affiliation(s)
- Carlos E. P. Leonardi
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Rodrigo A. Carrasco
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Fernanda C. F. Dias
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Eric M. Zwiefelhofer
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Gregg P. Adams
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Jaswant Singh
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
- * E-mail:
| |
Collapse
|
7
|
AKAD M, SOCOLOV R, COVALI R, STAN CD, CRAUCIUC E, POPOVICI D, STAN CI, AKAD F, SOCOLOV D. Kisspeptin Serum Levels in Patients with Endometriosis, New Research Pathways Regarding Female Infertility. MAEDICA 2022; 17:557-560. [PMID: 36540601 PMCID: PMC9720640 DOI: 10.26574/maedica.2022.17.3.557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Introduction:Endometriosis is defined by the presence of endometrial tissue outside the uterus, therefore leading to a chronic inflammatory reaction, adhesions development, scar tissue and a distorted pelvic female anatomy, most of the times leading to female infertility. Kisspeptin represents a neuropeptide thought to have an essential role in the reproductive functions of both female and male patients. Recently, positive correlations with kisspeptins were noticed in patients diagnosed with endometriosis. Materials and methods: Our study was performed between January 2021-March 2022 in "Elena Doamna" Clinical Hospital of Obstetrics and Gynecology Iasi, Romania. It was a prospective case-control study and included two groups of patients. Both groups consisted in female patients aged between 18 and 45 years, with a body mass index (BMI) between 18,5-30 kg/m2 and similar medical data. Patients in the study group had primary or secondary infertility and endometriosis, while the control group consisted of women with no reproductive issues who had healthy regular menstruations and at least one child. All patients agreed to participate in our study and signed the consent form. Clinical examination, pelvic ultrasound and hormonal dosages were performed. We tested the levels of LH, FSH, kisspeptin, estradiol, prolactin, testosterone, insulin and the glycemic levels in both groups during the follicular phase of their menstrual cycle. Results: We managed to enroll eight patients with endometriosis in the study group and an equal number of patients in the control group. There were significant differences between serum kisspeptin levels, but not also between other hormonal dosages. All patients in the study group had medical evidences of endometriomas but none of them had been subjected to laparoscopy. Conclusion:When considering our study, we regarded the first attribution given to kisspeptin, the one of a metastasis suppressor, and concluded that the high serum values of kisspeptin in patients with endometriosis represented a compensatory-adaptive mechanism needed to constrain future spread of endometriomas in early stages of this pathology.
Collapse
Affiliation(s)
- Mona AKAD
- “Gr. T. Popa” University of Medicine and Pharmacy, Iasi, Romania,“Elena Doamna” Clinical Hospital of Obstetrics and Gynecology, Iasi, Romania
| | - Razvan SOCOLOV
- “Gr. T. Popa” University of Medicine and Pharmacy, Iasi, Romania,“Elena Doamna” Clinical Hospital of Obstetrics and Gynecology, Iasi, Romania
| | - Roxana COVALI
- “Gr. T. Popa” University of Medicine and Pharmacy, Iasi, Romania,“Elena Doamna” Clinical Hospital of Obstetrics and Gynecology, Iasi, Romania
| | - Catalina Daniela STAN
- “Gr. T. Popa” University of Medicine and Pharmacy, Iasi, Romania,“Elena Doamna” Clinical Hospital of Obstetrics and Gynecology, Iasi, Romania
| | - Eduard CRAUCIUC
- “Gr. T. Popa” University of Medicine and Pharmacy, Iasi, Romania,“Elena Doamna” Clinical Hospital of Obstetrics and Gynecology, Iasi, Romania
| | - Diana POPOVICI
- “Gr. T. Popa” University of Medicine and Pharmacy, Iasi, Romania,“Elena Doamna” Clinical Hospital of Obstetrics and Gynecology, Iasi, Romania
| | | | - Fawzy AKAD
- “Gr. T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | - Demetra SOCOLOV
- “Gr. T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| |
Collapse
|
8
|
Camera M, Russo I, Zamboni V, Ammoni A, Rando S, Morellato A, Cimino I, Angelini C, Giacobini P, Oleari R, Amoruso F, Cariboni A, Franceschini I, Turco E, Defilippi P, Merlo GR. p140Cap Controls Female Fertility in Mice Acting via Glutamatergic Afference on Hypothalamic Gonadotropin-Releasing Hormone Neurons. Front Neurosci 2022; 16:744693. [PMID: 35237119 PMCID: PMC8884249 DOI: 10.3389/fnins.2022.744693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 01/20/2022] [Indexed: 11/13/2022] Open
Abstract
p140Cap, encoded by the gene SRCIN1 (SRC kinase signaling inhibitor 1), is an adaptor/scaffold protein highly expressed in the mouse brain, participating in several pre- and post-synaptic mechanisms. p140Cap knock-out (KO) female mice show severe hypofertility, delayed puberty onset, altered estrus cycle, reduced ovulation, and defective production of luteinizing hormone and estradiol during proestrus. We investigated the role of p140Cap in the development and maturation of the hypothalamic gonadotropic system. During embryonic development, migration of Gonadotropin-Releasing Hormone (GnRH) neurons from the nasal placode to the forebrain in p140Cap KO mice appeared normal, and young p140Cap KO animals showed a normal number of GnRH-immunoreactive (-ir) neurons. In contrast, adult p140Cap KO mice showed a significant loss of GnRH-ir neurons and a decreased density of GnRH-ir projections in the median eminence, accompanied by reduced levels of GnRH and LH mRNAs in the hypothalamus and pituitary gland, respectively. We examined the number of kisspeptin (KP) neurons in the rostral periventricular region of the third ventricle, the number of KP-ir fibers in the arcuate nucleus, and the number of KP-ir punctae on GnRH neurons but we found no significant changes. Consistently, the responsiveness to exogenous KP in vivo was unchanged, excluding a cell-autonomous defect on the GnRH neurons at the level of KP receptor or its signal transduction. Since glutamatergic signaling in the hypothalamus is critical for both puberty onset and modulation of GnRH secretion, we examined the density of glutamatergic synapses in p140Cap KO mice and observed a significant reduction in the density of VGLUT-ir punctae both in the preoptic area and on GnRH neurons. Our data suggest that the glutamatergic circuitry in the hypothalamus is altered in the absence of p140Cap and is required for female fertility.
Collapse
Affiliation(s)
- Mattia Camera
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Isabella Russo
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Valentina Zamboni
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Alessandra Ammoni
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Simona Rando
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Alessandro Morellato
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Irene Cimino
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Centre, Inserm U1172, Lille, France
- Metabolic Research Laboratories, Wellcome Trust–Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Costanza Angelini
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Paolo Giacobini
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Centre, Inserm U1172, Lille, France
| | - Roberto Oleari
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Federica Amoruso
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Anna Cariboni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Isabelle Franceschini
- Physiologie de la Reproduction et des Comportements, French National Centre for Scientific Research, French Institute of the Horse and Riding, French National Research Institute for Agriculture, Food and Environment, Université de Tours, Nouzilly, France
| | - Emilia Turco
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Paola Defilippi
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- *Correspondence: Paola Defilippi,
| | - Giorgio R. Merlo
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Giorgio R. Merlo,
| |
Collapse
|
9
|
Raúl Schmidt A, Ignacio Felipe Inserra P, Andrés Cortasa S, Proietto S, Fidel V, Halperin J, Daniel Vitullo A, Berta Dorfman V. Distribution of kisspeptin system and its relation with gonadotropin-releasing hormone in the hypothalamus of the South American plains vizcacha, Lagostomus maximus. Gen Comp Endocrinol 2022; 317:113974. [PMID: 34973969 DOI: 10.1016/j.ygcen.2021.113974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 12/21/2021] [Accepted: 12/26/2021] [Indexed: 11/20/2022]
Abstract
Kisspeptin (KISS), a key hormone involved in the regulation of the hypothalamic-pituitary-ovarian (HPO) axis, has been localized in the anteroventral periventricular (AVPV) nucleus and the neighboring rostral periventricular nucleus (PeVN), and in the arcuate (ARC) nucleus of the mammalian hypothalamus. In the ARC, the KISS neurons that co-express neurokinin B (NKB) and dynorphin A (Dyn) are named KNDy cells. The South American plains vizcacha is a rodent with peculiar reproductive traits. Around mid-pregnancy, vizcacha shows the reactivation of its HPO axis with the pulsatile release of gonadotropin-releasing hormone (GnRH) and luteinizing hormone (LH), an essential event for the success of gestation. Considering the role of KISS system in GnRH modulation, the aim of this work was to study their neuroanatomical distribution in adult vizcachas. AVPV showed sexual dimorphism with a significant smaller area in males (t-Test, p < 0.05), and KISS immunoreactivity was detected in somas and varicosities homogenously distributed in the AVPV with a concordant sex-related expression pattern. NKB and Dyn expression was also observed in cytoplasm of neurons scattered in the AVPV. Three subpopulations of neurons were detected in the AVPV: neurons expressing Dyn and NKB (DyNK cells), neurons expressing KISS and NKB (KiNK cells), and single NKB expressing neurons. Strikingly, KISS and Dyn were always expressed in different cells. In addition, in the ARC nucleus, KNDy cells were detected. On the other hand, KISS and GnRH expression was detected in different subpopulations of neurons, GnRH cells showed KISS receptor (KISSR or GPR-54) expression, and KISS immunoreactive afferent contacts were detected making close appositions onto somas and dendrites of GnRH cells. These results show similarities and differences between the KISS system in the hypothalamus of the vizcacha and other mammals, and constitute crucial observations about KISS and GnRH relation. Considering the peculiarity of HPO axis regulation in this species, the present work provides a neuroanatomical framework for the further elucidation of molecular mechanisms underlying GnRH expression and secretion.
Collapse
Affiliation(s)
- Alejandro Raúl Schmidt
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Pablo Ignacio Felipe Inserra
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Santiago Andrés Cortasa
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Sofía Proietto
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Victoria Fidel
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina
| | - Julia Halperin
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Alfredo Daniel Vitullo
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Verónica Berta Dorfman
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina.
| |
Collapse
|
10
|
Ikegami K, Watanabe Y, Nakamura S, Goto T, Inoue N, Uenoyama Y, Tsukamura H. Cellular and molecular mechanisms regulating the KNDy neuronal activities to generate and modulate GnRH pulse in mammals. Front Neuroendocrinol 2022; 64:100968. [PMID: 34808231 DOI: 10.1016/j.yfrne.2021.100968] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/18/2021] [Accepted: 11/15/2021] [Indexed: 12/30/2022]
Abstract
Accumulating findings during the past decades have demonstrated that the hypothalamic arcuate kisspeptin neurons are supposed to be responsible for pulsatile release of gonadotropin-releasing hormone (GnRH) to regulate gametogenesis and steroidogenesis in mammals. The arcuate kisspeptin neurons express neurokinin B (NKB) and dynorphin A (Dyn), thus, the neurons are also referred to as KNDy neurons. In the present article, we mainly focus on the cellular and molecular mechanisms underlying GnRH pulse generation, that is focused on the action of NKB and Dyn and an interaction between KNDy neurons and astrocytes to control GnRH pulse generation. Then, we also discuss the factors that modulate the activity of KNDy neurons and consequent pulsatile GnRH/LH release in mammals.
Collapse
Affiliation(s)
- Kana Ikegami
- Graduate School of Science, The University of Tokyo, Tokyo 113-0033, Japan
| | - Youki Watanabe
- Graduate School of Applied Life Science, Nippon Veterinary and Life Science University, Tokyo 180-8602, Japan
| | - Sho Nakamura
- Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Ehime 794-8555, Japan
| | - Teppei Goto
- RIKEN Center for Biosystems Dynamics Research, Hyogo 650-0047, Japan
| | - Naoko Inoue
- Graduate School of Bioagricultural Science, Nagoya University, Nagoya 464-8601, Japan
| | - Yoshihisa Uenoyama
- Graduate School of Bioagricultural Science, Nagoya University, Nagoya 464-8601, Japan
| | - Hiroko Tsukamura
- Graduate School of Bioagricultural Science, Nagoya University, Nagoya 464-8601, Japan.
| |
Collapse
|
11
|
Hypothalamic kisspeptin and kisspeptin receptors: Species variation in reproduction and reproductive behaviours. Front Neuroendocrinol 2022; 64:100951. [PMID: 34757093 DOI: 10.1016/j.yfrne.2021.100951] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/22/2021] [Accepted: 10/26/2021] [Indexed: 02/07/2023]
Abstract
Kisspeptin, encoded by the KISS1 gene, was first discovered as a potential metastasis suppressor gene. The prepro-kisspeptin precursor is cleaved into shorter mature bioactive peptides of varying sizes that bind to the G protein-coupled receptor GPR54 (=KISS1R). Over the last two decades, multiple types of Kiss and KissR genes have been discovered in mammalian and non-mammalian vertebrate species, but they are remarkably absent in birds. Kiss neuronal populations are distributed mainly in the hypothalamus. The KissRs are widely distributed in the brain, including the hypothalamic and non-hypothalamic regions, such as the hippocampus, amygdala, and habenula. The role of KISS1-KISS1R in humans and Kiss1-Kiss1R in rodents is associated with puberty, gonadal maturation, and the reproductive axis. However, recent gene deletion studies in zebrafish and medaka have provided controversial results, suggesting that the reproductive role of kiss is dispensable. This review highlights the evolutionary history, localisation, and significance of Kiss-KissR in reproduction and reproductive behaviours in mammalian and non-mammalian vertebrates.
Collapse
|
12
|
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: 4.0] [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
|
13
|
Ju M, Yang L, Zhu J, Chen Z, Zhang M, Yu J, Tian Z. MiR-664-2 impacts pubertal development in a precocious-puberty rat model through targeting the NMDA receptor-1†. Biol Reprod 2020; 100:1536-1548. [PMID: 30916745 DOI: 10.1093/biolre/ioz044] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 02/07/2019] [Accepted: 03/20/2019] [Indexed: 01/01/2023] Open
Abstract
Precocious puberty (PP) commonly results from premature activation of the hypothalamic-pituitary-gonadal axis (HPGA). Gonadotropin-releasing hormone (GnRH) is the initial trigger for HPGA activation and plays an important role in puberty onset. N-methyl-D-aspartate (NMDA) can promote pulsatile GnRH secretion and accelerates puberty onset. However, the mechanism of N-methyl-D-aspartate receptors (NMDARs) in PP pathogenesis remains obscure. We found that serum GnRH, luteinizing hormone (LH), follicle-stimulating hormone (FSH), estrogen (E2) levels, hypothalamic NMDAR1, and GnRH mRNA expression peaked at the vaginal opening (VO) day. Next, the hypothalamic NMDAR1 mRNA and protein levels in rats treated with danazol, a chemical commonly effecting on the reproductive system, were significantly increased at the VO day (postnatal day 24) compared to controls, accompanied by enhanced serum GnRH, LH, FSH, and E2 levels. Further, microRNA-664-2 (miR-664-2) was selected after bioinformatics analysis and approved in primary hypothalamic neurons, which binds to the 3'-untranslated regions of NMDAR1. Consistently, the miR-664-2 expression in hypothalamus of the Danazol group was decreased compared to Vehicle. Our results suggested that attenuated miR-664-2 might participate in PP pathogenesis through enhancing the NMDAR1 signaling.
Collapse
Affiliation(s)
- Minda Ju
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Institute of Acupuncture and Moxibustion Research, Academy of Integrative Medicine, Fudan University, Shanghai, China
| | - Liu Yang
- Shanghai Dunlu Biomedical Technology Co., Ltd, Shanghai, China
| | - Jing Zhu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Institute of Acupuncture and Moxibustion Research, Academy of Integrative Medicine, Fudan University, Shanghai, China
| | - Zhejun Chen
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Institute of Acupuncture and Moxibustion Research, Academy of Integrative Medicine, Fudan University, Shanghai, China
| | - Mizhen Zhang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Institute of Acupuncture and Moxibustion Research, Academy of Integrative Medicine, Fudan University, Shanghai, China
| | - Jin Yu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Institute of Acupuncture and Moxibustion Research, Academy of Integrative Medicine, Fudan University, Shanghai, China
| | - Zhanzhuang Tian
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Institute of Acupuncture and Moxibustion Research, Academy of Integrative Medicine, Fudan University, Shanghai, China
| |
Collapse
|
14
|
Glutamate Supply Reactivates Ovarian Function while Increases Serum Insulin and Triiodothyronine Concentrations in Criollo x Saanen-Alpine Yearlings' Goats during the Anestrous Season. Animals (Basel) 2020; 10:ani10020234. [PMID: 32024282 PMCID: PMC7070922 DOI: 10.3390/ani10020234] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 01/30/2020] [Accepted: 01/30/2020] [Indexed: 12/21/2022] Open
Abstract
The possible effect of glutamate supplementation upon ovarian reactivation and serum concentrations of insulin (INS) and triiodothyronine (T3) in anestrous yearling goats was evaluated. Goats (n = 32, 12 mo., 26° North, 1117 m) with a similar live weight (LW) and body condition score (BCS) were blood sampled twice per week for two weeks (2 × 1 week × 2 weeks) to confirm the anestrus status (<1 ng P4/mL; RIA). Thereafter, goats were randomly assigned to either 1) Glutamate (GLUT; n = 16, LW = 27.1 ± 1.09 kg, 3.5 ± 0.18 units, IV-supplemented with 7 mg of glutamate kg-1 LW), or 2) Control (CONT; n = 16; LW = 29.2 ± 1.09 kg; BCS = 3.5 ± 0.18, IV saline). During the treatment period, 16 goats (eight/group) were blood sampled twice per week for six weeks. Such serum samples (2 × 1 week × 6 weeks) were quantified by their P4 content to evaluate the ovarian-luteal activity, whereas a sample subset (1 × 1 week × 6 weeks) was used to quantify their INS & T3 content to evaluate their metabolic status. Neither LW (28.19 kg; p > 0.05) nor BCS (3.51 units; p > 0.05) differed between treatments. Goats depicting ovarian reactivation favored the GLUT group (50 vs. 12.5%; p < 0.05). Neither INS (1.72 ± 0.15 ng mL-1) nor T3 (2.32 ± 0.11 ng mL-1) differed between treatments, yet a treatment x time interaction regarding INS & T3 concentration across time favored (p < 0.05) the GLUT group. The results unveil exogenous glutamate as an interesting modulator not only of ovarian reactivation, but of metabolic hormone synthesis.
Collapse
|
15
|
Valle S, Das C, Meddle SL, Deviche P. The effect of food restriction on the regulation of gonadotropin-releasing hormone in male house finches (Haemorhous mexicanus). Gen Comp Endocrinol 2019; 282:113196. [PMID: 31163182 DOI: 10.1016/j.ygcen.2019.05.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 05/15/2019] [Accepted: 05/31/2019] [Indexed: 01/02/2023]
Abstract
Seasonal activation of the vertebrate hypothalamic-pituitary-gonadal (HPG) axis and gonadal development is initiated by gonadotropin-releasing hormone-I (GnRH) release from the hypothalamus. In photoperiodic species, the consistent annual change in photoperiod is the primary environmental signal affecting GnRH cell activity, including changes in the synthesis and secretion of this neuropeptide. Non-photoperiodic environmental cues such as energy availability also influence HPG axis activity, but the mechanisms mediating this influence, in particular on the GnRH system, are unclear. Understanding how the neuroendocrine system integrates environmental information is critical in determining the plasticity and adaptability of physiological responses to changing environments. The primary objective of this study was to investigate GnRH-mediated changes in HPG axis activity and gonadal development in response to energy availability in a wild bird. We hypothesized that negative energy balance inhibits HPG axis activity by affecting GnRH secretion. Moderate food restriction for several weeks in male house finches, Haemorhous mexicanus, decreased body condition and inhibited photoinduced testicular growth compared to birds fed ad libitum. Food restriction did not affect plasma luteinizing hormone (LH; a correlate of GnRH release) or plasma testosterone, but it enhanced the plasma LH response to an injection of the glutamatergic agonist, N-methyl-D-aspartate (NMDA). Thus, food restriction may decrease photoinduced HPG axis activation by acting centrally, in particular by attenuating the release of accumulated GnRH stores.
Collapse
Affiliation(s)
- Shelley Valle
- School of Life Sciences, Arizona State University, Tempe, AZ 85287-4501, USA.
| | - Chandrima Das
- School of Life Sciences, Arizona State University, Tempe, AZ 85287-4501, USA
| | - Simone L Meddle
- The Roslin Institute, The Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush, Midlothian EH25 9RG, Scotland, UK
| | - Pierre Deviche
- School of Life Sciences, Arizona State University, Tempe, AZ 85287-4501, USA
| |
Collapse
|
16
|
Plant TM. The neurobiological mechanism underlying hypothalamic GnRH pulse generation: the role of kisspeptin neurons in the arcuate nucleus. F1000Res 2019; 8. [PMID: 31297186 PMCID: PMC6600864 DOI: 10.12688/f1000research.18356.2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/20/2019] [Indexed: 01/21/2023] Open
Abstract
This review recounts the origins and development of the concept of the hypothalamic gonadotropin-releasing hormone (GnRH) pulse generator. It starts in the late 1960s when striking rhythmic episodes of luteinizing hormone secretion, as reflected by circulating concentrations of this gonadotropin, were first observed in monkeys and ends in the present day. It is currently an exciting time witnessing the application, primarily to the mouse, of contemporary neurobiological approaches to delineate the mechanisms whereby
Kiss1/NKB/Dyn (KNDy) neurons in the arcuate nucleus of the hypothalamus generate and time the pulsatile output of kisspeptin from their terminals in the median eminence that in turn dictates intermittent GnRH release and entry of this decapeptide into the primary plexus of the hypophysial portal circulation. The review concludes with an examination of questions that remain to be addressed.
Collapse
Affiliation(s)
- Tony M Plant
- Magee-Womens Research Institute, University of Pittsburgh, 204 Craft Avenue, Pittsburgh, PA 15213, USA
| |
Collapse
|
17
|
Plant TM. The neurobiological mechanism underlying hypothalamic GnRH pulse generation: the role of kisspeptin neurons in the arcuate nucleus. F1000Res 2019; 8:F1000 Faculty Rev-982. [PMID: 31297186 PMCID: PMC6600864 DOI: 10.12688/f1000research.18356.1] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/20/2019] [Indexed: 03/22/2024] Open
Abstract
This review recounts the origins and development of the concept of the hypothalamic gonadotropin-releasing hormone (GnRH) pulse generator. It starts in the late 1960s when striking rhythmic episodes of luteinizing hormone secretion, as reflected by circulating concentrations of this gonadotropin, were first observed in monkeys and ends in the present day. It is currently an exciting time witnessing the application, primarily to the mouse, of contemporary neurobiological approaches to delineate the mechanisms whereby Kiss1/NKB/Dyn (KNDy) neurons in the arcuate nucleus of the hypothalamus generate and time the pulsatile output of kisspeptin from their terminals in the median eminence that in turn dictates intermittent GnRH release and entry of this decapeptide into the primary plexus of the hypophysial portal circulation. The review concludes with an examination of questions that remain to be addressed.
Collapse
Affiliation(s)
- Tony M. Plant
- Magee-Womens Research Institute, University of Pittsburgh, 204 Craft Avenue, Pittsburgh, PA 15213, USA
| |
Collapse
|
18
|
Takács S, Bardóczi Z, Skrapits K, Göcz B, Váczi V, Maglóczky Z, Szűcs I, Rácz G, Matolcsy A, Dhillo WS, Watanabe M, Kádár A, Fekete C, Kalló I, Hrabovszky E. Post mortem single-cell labeling with DiI and immunoelectron microscopy unveil the fine structure of kisspeptin neurons in humans. Brain Struct Funct 2018; 223:2143-2156. [PMID: 29380121 DOI: 10.1007/s00429-018-1610-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 01/10/2018] [Indexed: 11/30/2022]
Abstract
Kisspeptin (KP) synthesizing neurons of the hypothalamic infundibular region are critically involved in the central regulation of fertility; these cells regulate pulsatile gonadotropin-releasing hormone (GnRH) secretion and mediate sex steroid feedback signals to GnRH neurons. Fine structural analysis of the human KP system is complicated by the use of post mortem tissues. To gain better insight into the neuroanatomy of the somato-dendritic cellular compartment, we introduced the diolistic labeling of immunohistochemically identified KP neurons using a gene gun loaded with the lipophilic dye, DiI. Confocal microscopic studies of primary dendrites in 100-µm-thick tissue sections established that 79.3% of KP cells were bipolar, 14.1% were tripolar, and 6.6% were unipolar. Primary dendrites branched sparsely, contained numerous appendages (9.1 ± 1.1 spines/100 µm dendrite), and received rich innervation from GABAergic, glutamatergic, and KP-containing terminals. KP neuron synaptology was analyzed with immunoelectron microscopy on perfusion-fixed specimens. KP axons established frequent contacts and classical synapses on unlabeled, and on KP-immunoreactive somata, dendrites, and spines. Synapses were asymmetric and the presynaptic structures contained round and regular synaptic vesicles, in addition to dense-core granules. Although immunofluorescent studies failed to detect vesicular glutamate transporter isoforms in KP axons, ultrastructural characteristics of synaptic terminals suggested use of glutamatergic, in addition to peptidergic, neurotransmission. In summary, immunofluorescent and DiI labeling of KP neurons in thick hypothalamic sections and immunoelectron microscopic studies of KP-immunoreactive neurons in brains perfusion-fixed shortly post mortem allowed us to identify previously unexplored fine structural features of KP neurons in the mediobasal hypothalamus of humans.
Collapse
Affiliation(s)
- Szabolcs Takács
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, 43 Szigony St., Budapest, 1083, Hungary
| | - Zsuzsanna Bardóczi
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
- School of PH.D. Studies, Semmelweis University, Budapest, Hungary
| | - Katalin Skrapits
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, 43 Szigony St., Budapest, 1083, Hungary
| | - Balázs Göcz
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, 43 Szigony St., Budapest, 1083, Hungary
| | - Viktória Váczi
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, 43 Szigony St., Budapest, 1083, Hungary
| | - Zsófia Maglóczky
- Human Brain Research Laboratory, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Iván Szűcs
- Department of Pathology, St. Borbála Hospital, Tatabanya, Hungary
| | - Gergely Rácz
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - András Matolcsy
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Waljit S Dhillo
- Department of Investigative Medicine, Hammersmith Hospital, Imperial College London, London, UK
| | - Masahiko Watanabe
- Department of Anatomy, Hokkaido University School of Medicine, Sapporo, Japan
| | - Andrea Kádár
- Laboratory of Integrative Neuroendocrinology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Csaba Fekete
- Laboratory of Integrative Neuroendocrinology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Tupper Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Imre Kalló
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
- Department of Neuroscience, Faculty of Information Technology, Pázmány Péter Catholic University, Budapest, Hungary
| | - Erik Hrabovszky
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, 43 Szigony St., Budapest, 1083, Hungary.
| |
Collapse
|
19
|
Bedenbaugh MN, O'Connell RC, Lopez JA, McCosh RB, Goodman RL, Hileman SM. Kisspeptin, gonadotrophin-releasing hormone and oestrogen receptor α colocalise with neuronal nitric oxide synthase neurones in prepubertal female sheep. J Neuroendocrinol 2018; 30:10.1111/jne.12560. [PMID: 29178496 PMCID: PMC5786465 DOI: 10.1111/jne.12560] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 11/02/2017] [Accepted: 11/21/2017] [Indexed: 01/07/2023]
Abstract
Puberty is a process that integrates multiple inputs ultimately resulting in an increase in gonadotrophin-releasing hormone (GnRH) secretion. Although kisspeptin neurones play an integral role in GnRH secretion and puberty onset, other systems are also likely important. One potential component is nitric oxide (NO), a gaseous neurotransmitter synthesised by nitric oxide synthase (NOS). The present study aimed to neuroanatomically characterise neuronal NOS (nNOS) in prepubertal female sheep and determine whether oestradiol exerts effects on this system. Luteinising hormone secretion was reduced by oestradiol treatment in prepubertal ovariectomised ewes. Neurones immunoreactive for nNOS were identified in several areas, with the greatest number present in the ventrolateral portion of the ventromedial hypothalamus, followed by the ventromedial hypothalamus, preoptic area (POA) and arcuate nucleus (ARC). Next, we determined whether nNOS neurones contained oestrogen receptor (ER)α and could potentially communicate oestradiol (E2 ) feedback to GnRH neurones. Neuronal NOS neurones contained ERα with the percentage of coexpression (12%-40%) depending upon the area analysed. We next investigated whether a neuroanatomical relationship existed between nNOS and kisspeptin or nNOS and GnRH neurones. A high percentage of kisspeptin neurones in the POA (79%) and ARC (98%) colocalised with nNOS. Kisspeptin close contacts were also associated with nNOS neurones. A greater number of close contacts were observed in the ARC than the POA. A high percentage of POA GnRH neurones (79%) also expressed nNOS, although no GnRH close contacts were observed onto nNOS neurones. Neither the numbers of nNOS neurones in the POA or hypothalamus, nor the percentage of nNOS coexpression with GnRH, kisspeptin or ERα were influenced by oestradiol. These experiments reveal that a neuroanatomical relationship exists between both nNOS and kisspeptin and nNOS and GnRH in prepubertal ewes. Therefore, nNOS may act both directly and indirectly to influence GnRH secretion in prepubertal sheep.
Collapse
Affiliation(s)
- M N Bedenbaugh
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV, USA
| | | | - J A Lopez
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV, USA
| | - R B McCosh
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV, USA
| | - R L Goodman
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV, USA
| | - S M Hileman
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
20
|
Chachlaki K, Garthwaite J, Prevot V. The gentle art of saying NO: how nitric oxide gets things done in the hypothalamus. Nat Rev Endocrinol 2017. [PMID: 28621341 DOI: 10.1038/nrendo.2017.69] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The chemical signalling molecule nitric oxide (NO), which freely diffuses through aqueous and lipid environments, subserves an array of functions in the mammalian central nervous system, such as the regulation of synaptic plasticity, blood flow and neurohormone secretion. In this Review, we consider the cellular and molecular mechanisms by which NO evokes short-term and long-term changes in neuronal activity. We also highlight recent studies showing that discrete populations of neurons that synthesize NO in the hypothalamus constitute integrative systems that support life by relaying metabolic and gonadal signals to the neuroendocrine brain, and thus gate the onset of puberty and adult fertility. The putative involvement and therapeutic potential of NO in the pathophysiology of brain diseases, for which hormonal imbalances during postnatal development could be risk factors, is also discussed.
Collapse
Affiliation(s)
- Konstantina Chachlaki
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Centre, UMR-S 1172, 1 place de Verdun, F-59000 Lille, France
- University of Lille, University Hospital Federations (FHU) 1,000 days for Health, School of Medicine, 1 place de Verdun, F-59000 Lille, France
| | - John Garthwaite
- The Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
| | - Vincent Prevot
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Centre, UMR-S 1172, 1 place de Verdun, F-59000 Lille, France
- University of Lille, University Hospital Federations (FHU) 1,000 days for Health, School of Medicine, 1 place de Verdun, F-59000 Lille, France
| |
Collapse
|
21
|
Abstract
Kisspeptins (KPs) and their receptor (GPR54 or KiSS1R) play a key-role in regulation of the hypothalamic-pituitary-gonadal axis and are therefore interesting targets for therapeutic interventions in the field of reproductive endocrinology. As dogs show a rapid and robust LH response after the administration of KP10, they can serve as a good animal model for research concerning KP signaling. The aims of the present study were to test the antagonistic properties of KP analogs p234, p271, p354, and p356 in vitro, by determining the intracellular Ca2+ response of CHEM1 cells that stably express human GPR54, and to study the in vivo effects of these peptides on basal plasma LH concentration and the KP10-induced LH response in female dogs. Exposure of the CHEM1 cells to KP-10 resulted in a clear Ca2+ response. P234, p271, p354, and p356 did not prevent or lower the KP10-induced Ca2+ response. Moreover, the in vivo studies in the dogs showed that none of these supposed antagonists lowered the basal plasma LH concentration and none of the peptides lowered the KP10-induced LH response. In conclusion, p234, p271, p354, and p356 had no antagonistic effects in vitro nor any effect on basal and kisspeptin-stimulated plasma LH concentration in female dogs.
Collapse
|
22
|
d'Anglemont de Tassigny X, Jayasena C, Murphy KG, Dhillo WS, Colledge WH. Mechanistic insights into the more potent effect of KP-54 compared to KP-10 in vivo. PLoS One 2017; 12:e0176821. [PMID: 28464043 PMCID: PMC5413024 DOI: 10.1371/journal.pone.0176821] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 04/18/2017] [Indexed: 01/03/2023] Open
Abstract
Kisspeptins regulate the mammalian reproductive axis by stimulating release of gonadotrophin releasing hormone (GnRH). Different length kisspeptins (KP) are found of 54, 14, 13 or 10 amino-acids which share a common C-terminal 10-amino acid sequence. KP-54 and KP-10 have been widely used to stimulate the reproductive axis but data suggest that KP-54 and KP-10 are not equally effective at eliciting reproductive hormone secretion after peripheral delivery. To confirm this, we analysed the effect of systemic administration of KP-54 or KP-10 on luteinizing hormone (LH) secretion into the bloodstream of male mice. Plasma LH measurements 10 min or 2 hours after kisspeptin injection showed that KP-54 can sustain LH release far longer than KP-10, suggesting a differential mode of action of the two peptides. To investigate the mechanism for this, we evaluated the pharmacokinetics of the two peptides in vivo and their potential to cross the blood brain barrier (BBB). We found that KP-54 has a half-life of ~32 min in the bloodstream, while KP-10 has a half-life of ~4 min. To compensate for this difference in half-life, we repeated injections of KP-10 every 10 min over 1 hr but failed to reproduce the sustained rise in LH observed after a single KP-54 injection, suggesting that the failure of KP-10 to sustain LH release may not just be related to peptide clearance. We tested the ability of peripherally administered KP-54 and KP-10 to activate c-FOS in GnRH neurons behind the blood brain barrier (BBB) and found that only KP-54 could do this. These data are consistent with KP-54 being able to cross the BBB and suggest that KP10 may be less able to do so.
Collapse
Affiliation(s)
- Xavier d'Anglemont de Tassigny
- Reproductive Physiology Group, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Channa Jayasena
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London at Hammersmith Campus, Commonwealth Building, London, United Kingdom
| | - Kevin G. Murphy
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London at Hammersmith Campus, Commonwealth Building, London, United Kingdom
| | - Waljit S. Dhillo
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London at Hammersmith Campus, Commonwealth Building, London, United Kingdom
| | - William H. Colledge
- Reproductive Physiology Group, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
- * E-mail:
| |
Collapse
|
23
|
Rabconnectin-3α is required for the morphological maturation of GnRH neurons and kisspeptin responsiveness. Sci Rep 2017; 7:42463. [PMID: 28209974 PMCID: PMC5314327 DOI: 10.1038/srep42463] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 01/11/2017] [Indexed: 12/18/2022] Open
Abstract
A few hundred hypothalamic neurons form a complex network that controls reproduction in mammals by secreting gonadotropin-releasing hormone (GnRH). Timely postnatal changes in GnRH secretion are essential for pubertal onset. During the juvenile period, GnRH neurons undergo morphological remodeling, concomitantly achieving an increased responsiveness to kisspeptin, the main secretagogue of GnRH. However, the link between GnRH neuron activity and their morphology remains unknown. Here, we show that brain expression levels of Dmxl2, which encodes the vesicular protein rabconnectin-3α, determine the capacity of GnRH neurons to be activated by kisspeptin and estradiol. We also demonstrate that Dmxl2 expression levels control the pruning of GnRH dendrites, highlighting an unexpected role for a vesicular protein in the maturation of GnRH neuronal network. This effect is mediated by rabconnectin-3α in neurons or glial cells afferent to GnRH neurons. The widespread expression of Dmxl2 in several brain areas raises the intriguing hypothesis that rabconnectin-3α could be involved in the maturation of other neuronal populations.
Collapse
|
24
|
Messina A, Langlet F, Chachlaki K, Roa J, Rasika S, Jouy N, Gallet S, Gaytan F, Parkash J, Tena-Sempere M, Giacobini P, Prevot V. A microRNA switch regulates the rise in hypothalamic GnRH production before puberty. Nat Neurosci 2016; 19:835-44. [DOI: 10.1038/nn.4298] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 04/01/2016] [Indexed: 12/12/2022]
|
25
|
Meczekalski B, Katulski K, Podfigurna-Stopa A, Czyzyk A, Genazzani AD. Spontaneous endogenous pulsatile release of kisspeptin is temporally coupled with luteinizing hormone in healthy women. Fertil Steril 2016; 105:1345-1350.e2. [PMID: 26859129 DOI: 10.1016/j.fertnstert.2016.01.029] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 12/24/2015] [Accepted: 01/20/2016] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To evaluate the presence of a spontaneous pulsatile release of kisspeptin and whether it is temporally coupled to LH pulses. DESIGN Experimental study. SETTING Academic medical center. PATIENT(S) Thirty young healthy eumenorrheic women aged 20-37 years were included in the study group. All subjects were white women admitted to the Department of Gynecological Endocrinology, Poznan University of Medical Sciences, Poznan, Poland. INTERVENTION(S) Kisspeptin, FSH, LH, E2, PRL, and insulin were evaluated in all subjects at baseline. MAIN OUTCOME MEASURE(S) All women underwent a pulsatility study measuring LH and kisspeptin plasma concentrations to assess the spontaneous episodic secretion of both hormones, sampling every 10 minutes for 2 hours from 9:00 to 11:00 a.m. for a total of 12 blood samples. Detection and specific concordance (SC) algorithms were used to detect pulses and their concordance. RESULT(S) A significant endogenous secretory pattern was demonstrated for both LH and kisspeptin over the 2-hour duration of the study (2.4 ± 0.1 peaks/2 h). The computation of the SC index showed for the first time that kisspeptin and LH are cosecreted and temporally coupled at time "0," and their peaks occur at the same point in time. CONCLUSION(S) The present study provides evidence supporting the hypothesis that kisspeptin is highly relevant in the regulation and modulation of reproductive functions in humans.
Collapse
Affiliation(s)
- Blazej Meczekalski
- Department of Gynecological Endocrinology, Poznan University of Medical Sciences, Poznan, Poland.
| | - Krzysztof Katulski
- Department of Gynecological Endocrinology, Poznan University of Medical Sciences, Poznan, Poland
| | | | - Adam Czyzyk
- Department of Gynecological Endocrinology, Poznan University of Medical Sciences, Poznan, Poland
| | - Alessandro D Genazzani
- Department of Obstetrics and Gynecology, Gynecological Endocrinology Center, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
26
|
Büdefeld T, Tobet S, Majdic G. The Influence of Gonadal Steroid Hormones on Immunoreactive Kisspeptin in the Preoptic Area and Arcuate Nucleus of Developing Agonadal Mice with a Genetic Disruption of Steroidogenic Factor 1. Neuroendocrinology 2016; 103:248-58. [PMID: 26138474 PMCID: PMC4696913 DOI: 10.1159/000437166] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 06/22/2015] [Indexed: 11/19/2022]
Abstract
Kisspeptin, a regulator of reproductive function and puberty in mammals, is expressed in the rostral (anteroventral) periventricular nucleus (AVPV) and arcuate nucleus (Arc), and its expression is at least partially regulated by estradiol in rodents. The aim of the present study was to determine contributions of genetic factors and gonadal steroid hormones to the sexual differentiation of kisspeptin-immunoreactive (kisspeptin-ir) cell populations in the AVPV and Arc during postnatal development using agonadal steroidogenic factor 1 (SF-1) knockout (KO) mice. To examine the effects of gonadal hormones on pubertal development of kisspeptin neurons, SF-1 KO mice were treated with estradiol benzoate (EB) from postnatal day (P)25 to P36, and their brains were examined at P36. No sex differences were observed in the SF-1 KO mice during postnatal development and after treatment with EB - which failed to increase the number of kisspeptin-ir cells at P36 to the levels found in wild-type (WT) control females. This suggests that specific time periods of estradiol actions or other factors are needed for sexual differentiation of the pattern of immunoreactive kisspeptin in the AVPV. Kisspeptin immunoreactivity in the Arc was significantly higher in gonadally intact WT and SF-1 KO females than in male mice at P36 during puberty. Further, in WT and SF-1 KO females, but not in males, adult levels were reached at P36. This suggests that maturation of the kisspeptin system in the Arc differs between sexes and is regulated by gonad-independent mechanisms.
Collapse
Affiliation(s)
- Tomaz Büdefeld
- Centre for Animal Genomics, Veterinary Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Stuart Tobet
- Department of Biomedical Sciences and School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
| | - Gregor Majdic
- Centre for Animal Genomics, Veterinary Faculty, University of Ljubljana, Ljubljana, Slovenia
- Institute of Physiology, Medical School, University of Maribor, Maribor, Slovenia
- Corresponding author and person to whom proofs and reprint requests should be addressed: Gregor Majdic; Center for Animal Genomics, Veterinary Faculty, University of Ljubljana, Slovenia-1000 Ljubljana; Phone: 0038614779210, Fax: 0038612832243,
| |
Collapse
|
27
|
Abstract
The endocrine hypothalamus constitutes those cells which project to the median eminence and secrete neurohormones into the hypophysial portal blood to act on cells of the anterior pituitary gland. The entire endocrine system is controlled by these peptides. In turn, the hypothalamic neuroendocrine cells are regulated by feedback signals from the endocrine glands and other circulating factors. The neuroendocrine cells are found in specific regions of the hypothalamus and are regulated by afferents from higher brain centers. Integrated function is clearly complex and the networks between and amongst the neuroendocrine cells allows fine control to achieve homeostasis. The entry of hormones and other factors into the brain, either via the cerebrospinal fluid or through fenestrated capillaries (in the basal hypothalamus) is important because it influences the extent to which feedback regulation may be imposed. Recent evidence of the passage of factors from the pars tuberalis and the median eminence casts a new layer in our understanding of neuroendocrine regulation. The function of neuroendocrine cells and the means by which pulsatile secretion is achieved is best understood for the close relationship between gonadotropin releasing hormone and luteinizing hormone, which is reviewed in detail. The secretion of other neurohormones is less rigid, so the relationship between hypothalamic secretion and the relevant pituitary hormones is more complex.
Collapse
Affiliation(s)
- I J Clarke
- Monash University, Department of Physiology, Clayton, Australia
| |
Collapse
|
28
|
Shamas S, Khan SUH, Khan MY, Shabbir N, Zubair H, Shafqat S, Wahab F, Shahab M. Fasting induced kisspeptin signaling suppression is regulated by glutamate mediated cues in adult male rhesus macaque (Macaca mulatta). Neuropeptides 2015; 52:39-45. [PMID: 26138506 DOI: 10.1016/j.npep.2015.06.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 06/09/2015] [Accepted: 06/10/2015] [Indexed: 11/18/2022]
Abstract
Kisspeptin signaling is suppressed by short term fasting. It has been reported that hypothalamic Kiss1 and Kiss1r mRNA expression decreased after 48h of fasting in male rhesus monkey. But the mechanism involved in the reduction of kisspeptin signaling after 48h of fasting is unknown. Recent studies have suggested the role of afferent excitatory and inhibitory pathways in the regulation of kisspeptin neurons. Therefore, this study was designed to observe the changes in the glutamate and GABA signaling during fed and 48h fasting states by performing immunofluorescence to examine the interaction of kisspeptin neurons with NR1 subunit of NMDA receptors and by performing SYBR green qRT-PCR to measure and quantify the levels of Kiss1, Kiss1r, NR1 and GAD67 mRNA in the POA and MBH of adult male rhesus macaque (Macaca mulatta) during 48h of fasting (n=2) and fed ad libitum (n=2). Plasma testosterone (p<0.05) and blood glucose levels were significantly (p<0.001) decreased after short term fasting. Our results clearly showed that expression of hypothalamic Kiss1, Kiss1r and NR1 mRNA was significantly (p<0.05) reduced in adult male rhesus monkeys which were fasted for 48h as compared to those which were fed ad libitum. There was no clear difference in the GAD67 mRNA contents between the two groups. Number of kisspeptin neurons and the interactions of kisspeptin neurons with NR1 were significantly (p<0.05) reduced after 48h fasting. These observations suggest that decreased kisspeptin signaling during fasting may occur due to reduction in glutamatergic inputs to kisspeptin neurons. Our results also suggest that fasting induced suppression of kisspeptin signaling is not mediated through GABAergic neurons.
Collapse
Affiliation(s)
- Shazia Shamas
- Laboratory of Reproductive Neuroendocrinology, Department of Animal Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Saeed-Ul-Hassan Khan
- Department of Pathology, Shaheed Zulfiqar Ali Bhutto Medical University, Islamabad, Pakistan
| | - Muhammad Yousaf Khan
- Department of Pathology, Shaheed Zulfiqar Ali Bhutto Medical University, Islamabad, Pakistan
| | - Nadia Shabbir
- Laboratory of Reproductive Neuroendocrinology, Department of Animal Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Hira Zubair
- Laboratory of Reproductive Neuroendocrinology, Department of Animal Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Saira Shafqat
- Laboratory of Reproductive Neuroendocrinology, Department of Animal Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Fazal Wahab
- Laboratory of Reproductive Neuroendocrinology, Department of Animal Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Muhammad Shahab
- Laboratory of Reproductive Neuroendocrinology, Department of Animal Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| |
Collapse
|
29
|
Jayasena CN, Abbara A, Narayanaswamy S, Comninos AN, Ratnasabapathy R, Bassett P, Mogford JT, Malik Z, Calley J, Ghatei MA, Bloom SR, Dhillo WS. Direct comparison of the effects of intravenous kisspeptin-10, kisspeptin-54 and GnRH on gonadotrophin secretion in healthy men. Hum Reprod 2015; 30:1934-41. [PMID: 26089302 PMCID: PMC4507333 DOI: 10.1093/humrep/dev143] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 05/22/2015] [Indexed: 12/16/2022] Open
Abstract
STUDY QUESTION How potently does the novel hypothalamic stimulator of reproduction, kisspeptin, increase gonadotrophin secretion when compared with GnRH in healthy men? SUMMARY ANSWER At the doses tested, intravenous administration of either of two major kisspeptin isoforms, kisspeptin-10 and -54, was associated with similar levels of gonadotrophin secretion in healthy men; however, GnRH was more potent when compared with either kisspeptin isoform. WHAT IS KNOWN ALREADY Kisspeptin-10 and -54 are naturally occurring hormones in the kisspeptin peptide family which potently stimulates endogenous GnRH secretion from the hypothalamus, so have the potential to treat patients with reproductive disorders. Rodent studies suggest that kisspeptin-54 is more potent when compared with kisspepitn-10; however, their effects have not previously been directly compared in humans, or compared with direct pituitary stimulation of gonadotrophin secretion using GnRH. STUDY DESIGN, SIZE AND DURATION A single-blinded placebo controlled physiological study was performed from January to December 2013. Local ethical approval was granted, and five participants were recruited to each dosing group. PARTICIPANTS/MATERIALS, SETTING, METHODS Healthy men were administered vehicle, kisspeptin-10, kisspeptin-54 and GnRH intravenously for 3 h on different study days. Each hormone was administered at 0.1, 0.3 and 1.0 nmol/kg/h doses (n = 5 subjects per group). Regular blood sampling was conducted throughout the study to measure LH and FSH. Study visits were conducted at least a week apart. MAIN RESULTS AND THE ROLE OF CHANCE Serum LH and FSH levels were ∼3-fold higher during GnRH infusion when compared with kisspeptin-10 and ∼2-fold higher when compared with kisspeptin-54 [mean area under the curve serum LH during infusion (in hours times international units per litre, h.IU/l): 10.81 ± 1.73, 1.0 nmol/kg/h kisspeptin-10; 14.43 ± 1.27, 1.0 nmol/kg/h kisspeptin-54; 34.06 ± 5.18, 1.0 nmol/kg/h GnRH, P < 0.001 versus kisspeptin-10, P < 0.01 versus kisspeptin-54]. LIMITATIONS, REASONS FOR CAUTION This study had a small sample size. WIDER IMPLICATIONS OF THE FINDINGS Kisspeptin offers a novel means of stimulating the reproductive axis. Our data suggest that kisspeptin stimulates gonadotrophin secretion less potently when compared with GnRH; however, kisspeptin may stimulate gonadotrophins in a more physiological manner when compared with current therapies. Kisspeptin is emerging as a future therapeutic agent, so it is important to establish which kisspeptin hormones could be used to treat patients with infertility. Results of this study suggest that either isoform has similar effects on reproductive hormone secretion in healthy men when administered intravenously. STUDY FUNDING/COMPETING INTERESTS This work is funded by grants from the MRC and NIHR and is supported by the NIHR Imperial Biomedical Research Centre Funding Scheme. C.N.J. is supported by an NIHR Clinical Lectureship. A.A. is supported by Wellcome Trust Research Training Fellowships. A.N.C. is supported by Wellcome Trust Translational Medicine Training Fellowship. W.S.D. is supported by an NIHR Career Development Fellowship.
Collapse
Affiliation(s)
- C N Jayasena
- Department of Investigative Medicine, Imperial College London, Hammersmith Hospital, 6th Floor, Commonwealth Building, London W12 0NN, UK
| | - A Abbara
- Department of Investigative Medicine, Imperial College London, Hammersmith Hospital, 6th Floor, Commonwealth Building, London W12 0NN, UK
| | - S Narayanaswamy
- Department of Investigative Medicine, Imperial College London, Hammersmith Hospital, 6th Floor, Commonwealth Building, London W12 0NN, UK
| | - A N Comninos
- Department of Investigative Medicine, Imperial College London, Hammersmith Hospital, 6th Floor, Commonwealth Building, London W12 0NN, UK
| | - R Ratnasabapathy
- Department of Investigative Medicine, Imperial College London, Hammersmith Hospital, 6th Floor, Commonwealth Building, London W12 0NN, UK
| | - P Bassett
- Statsconsultancy Ltd, 40 Longwood Lane, Amersham, Bucks HP7 9EN, UK
| | - J T Mogford
- Department of Investigative Medicine, Imperial College London, Hammersmith Hospital, 6th Floor, Commonwealth Building, London W12 0NN, UK
| | - Z Malik
- Department of Investigative Medicine, Imperial College London, Hammersmith Hospital, 6th Floor, Commonwealth Building, London W12 0NN, UK
| | - J Calley
- Department of Investigative Medicine, Imperial College London, Hammersmith Hospital, 6th Floor, Commonwealth Building, London W12 0NN, UK
| | - M A Ghatei
- Department of Investigative Medicine, Imperial College London, Hammersmith Hospital, 6th Floor, Commonwealth Building, London W12 0NN, UK
| | - S R Bloom
- Department of Investigative Medicine, Imperial College London, Hammersmith Hospital, 6th Floor, Commonwealth Building, London W12 0NN, UK
| | - W S Dhillo
- Department of Investigative Medicine, Imperial College London, Hammersmith Hospital, 6th Floor, Commonwealth Building, London W12 0NN, UK
| |
Collapse
|
30
|
Ezzat A, Pereira A, Clarke IJ. Kisspeptin is a component of the pulse generator for GnRH secretion in female sheep but not the pulse generator. Endocrinology 2015; 156:1828-37. [PMID: 25710282 DOI: 10.1210/en.2014-1756] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We tested the hypothesis that kisspeptin cells constitute the "pulse generator" for GnRH secretion. In ewes, we determined whether iv administered kisspeptin elicits a secretory pulse of LH in anaesthetized, sex-steroid suppressed ovariectomized ewes. A response was seen in both anaesthetized and conscious animals, which was not associated with induction of c-Fos labeling in GnRH cells, supporting the notion that kisspeptin acts on the neurosecretory GnRH terminals. Response was lower in the anaesthetized animals, suggesting that some nonkisspeptin elements may be involved in GnRH responses. Microinjection of kisspeptin (100 nmol) into the median eminence of conscious ewes elicited a pulse of LH, indicating that kisspeptin acts at this level to cause GnRH secretion. To determine which cells are activated at the time of GnRH secretion, we blood sampled 18 ewes during the luteal phase of the estrous cycle and harvested brains after 3 hours. Three of these ewes displayed a pulse of LH within 30 minutes of euthanasia. An increase in c-Fos labeling was seen in kisspeptin and glutamate cells of the arcuate nucleus but not in GnRH neurons, preoptic kisspeptin neurons, or preoptic glutamate neurons. Immunohistochemistry in 4 hypothalami showed that 72% of arcuate kisspeptin cells receive glutamatergic input. These data support the concept that the kisspeptin cells of the arcuate nucleus drive pulsatile secretion of GnRH at the level of the median eminence, but this may involve "upstream" input from glutamate cells. We conclude that the pulse generator for GnRH secretion involves more than 1 element.
Collapse
Affiliation(s)
- Ahmed Ezzat
- Department of Physiology, Monash University, Clayton, Victoria 3800, Australia
| | | | | |
Collapse
|
31
|
Comninos AN, Anastasovska J, Sahuri-Arisoylu M, Li X, Li S, Hu M, Jayasena CN, Ghatei MA, Bloom SR, Matthews PM, O'Byrne KT, Bell JD, Dhillo WS. Kisspeptin signaling in the amygdala modulates reproductive hormone secretion. Brain Struct Funct 2015; 221:2035-47. [PMID: 25758403 PMCID: PMC4853463 DOI: 10.1007/s00429-015-1024-9] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 03/02/2015] [Indexed: 12/27/2022]
Abstract
Kisspeptin (encoded by KISS1) is a crucial activator of reproductive function. The role of kisspeptin has been studied extensively within the hypothalamus but little is known about its significance in other areas of the brain. KISS1 and its cognate receptor are expressed in the amygdala, a key limbic brain structure with inhibitory projections to hypothalamic centers involved in gonadotropin secretion. We therefore hypothesized that kisspeptin has effects on neuronal activation and reproductive pathways beyond the hypothalamus and particularly within the amygdala. To test this, we mapped brain neuronal activity (using manganese-enhanced MRI) associated with peripheral kisspeptin administration in rodents. We also investigated functional relevance by measuring the gonadotropin response to direct intra-medial amygdala (MeA) administration of kisspeptin and kisspeptin antagonist. Peripheral kisspeptin administration resulted in a marked decrease in signal intensity in the amygdala compared to vehicle alone. This was associated with an increase in luteinizing hormone (LH) secretion. In addition, intra-MeA administration of kisspeptin resulted in increased LH secretion, while blocking endogenous kisspeptin signaling within the amygdala by administering intra-MeA kisspeptin antagonist decreased both LH secretion and LH pulse frequency. We provide evidence for the first time that neuronal activity within the amygdala is decreased by peripheral kisspeptin administration and that kisspeptin signaling within the amygdala contributes to the modulation of gonadotropin release and pulsatility. Our data suggest that kisspeptin is a 'master regulator' of reproductive physiology, integrating limbic circuits with the regulation of gonadotropin-releasing hormone neurons and reproductive hormone secretion.
Collapse
Affiliation(s)
- Alexander N Comninos
- Department of Investigative Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - Jelena Anastasovska
- Metabolic and Molecular Imaging Group, MRC Clinical Science Centre, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - Meliz Sahuri-Arisoylu
- Metabolic and Molecular Imaging Group, MRC Clinical Science Centre, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - Xiaofeng Li
- Division of Women's Health, School of Medicine, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Shengyun Li
- Division of Women's Health, School of Medicine, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Minghan Hu
- Division of Women's Health, School of Medicine, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Channa N Jayasena
- Department of Investigative Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - Mohammad A Ghatei
- Department of Investigative Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - Stephen R Bloom
- Department of Investigative Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - Paul M Matthews
- Division of Brain Sciences, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Kevin T O'Byrne
- Division of Women's Health, School of Medicine, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Jimmy D Bell
- Metabolic and Molecular Imaging Group, MRC Clinical Science Centre, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - Waljit S Dhillo
- Department of Investigative Medicine, Imperial College London, 6th Floor Commonwealth Building, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
32
|
Uenoyama Y, Nakamura S, Hayakawa Y, Ikegami K, Watanabe Y, Deura C, Minabe S, Tomikawa J, Goto T, Ieda N, Inoue N, Sanbo M, Tamura C, Hirabayashi M, Maeda KI, Tsukamura H. Lack of pulse and surge modes and glutamatergic stimulation of luteinising hormone release in Kiss1 knockout rats. J Neuroendocrinol 2015; 27:187-97. [PMID: 25582792 DOI: 10.1111/jne.12257] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Revised: 01/05/2015] [Accepted: 01/05/2015] [Indexed: 12/11/2022]
Abstract
Kisspeptin, encoded by the Kiss1 gene, has attracted attention as a key candidate neuropeptide in controlling puberty and reproduction via regulation of gonadotrophin-releasing hormone (GnRH) secretion in mammals. Pioneer studies with Kiss1 or its cognate receptor Gpr54 knockout (KO) mice showed the indispensable role of kisspeptin-GPR54 signalling in the control of animal reproduction, although detailed analyses of gonadotrophin secretion, especially pulsatile and surge-mode of luteinising hormone (LH) secretion, were limited. Thus, in the present study, we have generated Kiss1 KO rats aiming to evaluate a key role of kisspeptin in governing reproduction via pulse and surge modes of GnRH/LH secretion. Kiss1 KO male and female rats showed a complete suppression of pulsatile LH secretion, which is responsible for folliculogenesis and spermatogenesis, and an absence of puberty and atrophic gonads. Kiss1 KO female rats showed no spontaneous LH/follicle-stimulating hormone surge and an oestrogen-induced LH surge, suggesting that the GnRH surge generation system, which is responsible for ovulation, does not function without kisspeptin. Furthermore, challenge of major stimulatory neurotransmitters, such as monosodium glutamate, NMDA and norepinephrine, failed to stimulate LH secretion in Kiss1 KO rats, albeit they stimulated LH release in wild-type controls. Taken together, the results of the present study confirm that kisspeptin plays an indispensable role in generating two modes (pulse and surge) of GnRH/gonadotrophin secretion to regulate puberty onset and normal reproductive performance. In addition, the present study suggests that kisspeptin neurones play a critical role as a hub integrating major stimulatory neural inputs to GnRH neurones, using newly established Kiss1 KO rats, which serve as a useful model for detailed analysis of hormonal profiles.
Collapse
Affiliation(s)
- Y Uenoyama
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Beymer M, Negrón AL, Yu G, Wu S, Mayer C, Lin RZ, Boehm U, Acosta-Martínez M. Kisspeptin cell-specific PI3K signaling regulates hypothalamic kisspeptin expression and participates in the regulation of female fertility. Am J Physiol Endocrinol Metab 2014; 307:E969-82. [PMID: 25269483 PMCID: PMC4254985 DOI: 10.1152/ajpendo.00385.2014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hypothalamic kisspeptin neurons integrate and translate cues from the internal and external environments that regulate gonadotropin-releasing hormone (GnRH) secretion and maintain fertility in mammals. However, the intracellular signaling pathways utilized to translate such information into changes in kisspeptin expression, release, and ultimately activation of the kisspeptin-receptive GnRH network have not yet been identified. PI3K is an important signaling node common to many peripheral factors known to regulate kisspeptin expression and GnRH release. We investigated whether PI3K signaling regulates hypothalamic kisspeptin expression, pubertal development, and adult fertility in mice. We generated mice with a kisspeptin cell-specific deletion of the PI3K catalytic subunits p110α and p110β (kiss-p110α/β-KO). Using in situ hybridization, we examined Kiss1 mRNA expression in gonad-intact, gonadectomized (Gdx), and Gdx + steroid-replaced mice. Kiss1 cell number in the anteroventral periventricular hypothalamus (AVPV) was significantly reduced in intact females but not in males. In contrast, compared with WT and regardless of steroid hormone status, Kiss1 cell number was lower in the arcuate (ARC) of kiss-p110α/β-KO males, but it was unaffected in females. Both intact Kiss-p110α/β-KO males and females had reduced ARC kisspeptin-immunoreactive (IR) fibers compared with WT animals. Adult kiss-p110α/β-KO males had significantly lower circulating luteinizing hormone (LH) levels, whereas pubertal development and fertility were unaffected in males. Kiss-p110α/β-KO females exhibited a reduction in fertility despite normal pubertal development, LH levels, and estrous cyclicity. Our data show that PI3K signaling is important for the regulation of hypothalamic kisspeptin expression and contributes to normal fertility in females.
Collapse
Affiliation(s)
- Matthew Beymer
- Department of Physiology and Biophysics, Stony Brook University Medical Center, Stony Brook, New York; Graduate Program in Genetics, Stony Brook University, Stony Brook, New York
| | - Ariel L Negrón
- Department of Physiology and Biophysics, Stony Brook University Medical Center, Stony Brook, New York; Graduate Program in Neuroscience, Stony Brook University, Stony Brook, New York
| | - Guiqin Yu
- Department of Physiology and Biophysics, Stony Brook University Medical Center, Stony Brook, New York
| | - Samuel Wu
- Department of Physiology and Biophysics, Stony Brook University Medical Center, Stony Brook, New York
| | - Christian Mayer
- Department of Pharmacology and Toxicology, University of Saarland School of Medicine, Homburg, Germany
| | - Richard Z Lin
- Department of Physiology and Biophysics, Stony Brook University Medical Center, Stony Brook, New York; Institute of Molecular Cardiology, Stony Brook, New York; and Veterans Affairs Medical Center, Northport, New York
| | - Ulrich Boehm
- Department of Pharmacology and Toxicology, University of Saarland School of Medicine, Homburg, Germany
| | - Maricedes Acosta-Martínez
- Department of Physiology and Biophysics, Stony Brook University Medical Center, Stony Brook, New York;
| |
Collapse
|
34
|
Liu X, Herbison AE. RF9 excitation of GnRH neurons is dependent upon Kiss1r in the adult male and female mouse. Endocrinology 2014; 155:4915-24. [PMID: 25322463 DOI: 10.1210/en.2014-1517] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The neuropeptide FF receptor antagonist 1-adamantanecarbonyl-Arg-Phe-NH2 trifluoroacetate salt (RF9) has been found to be a remarkably potent activator of gonadotropin secretion in mammals. However, the mechanism of RF9 action on the reproductive axis is unknown. Using acute brain slice electrophysiology in genetically modified mouse models, we have investigated the possibility that RF9 may activate GnRH neurons. In transgenic GnRH-GFP male and female mice, RF9 was found to exert potent, dose-dependent, stimulatory effects on the firing rate of approximately 70% of GnRH neurons. These effects occurred directly on GnRH neurons and were independent of fast amino acid transmission. To assess RF9's action as an neuropeptide FF receptor antagonist at the GnRH neuron, its ability to antagonize the inhibitory effects of RFamide-related peptide-3 on GnRH neuron firing was examined. RF9 exhibited variable ability to prevent the inhibitory effects of RFamide-related peptide-3 on GnRH neurons. Whole-cell recordings from GnRH neurons showed that RF9 generated an inward current in GnRH neurons reminiscent of that evoked by kisspeptin. We therefore examined RF9 actions in kisspeptin receptor knockout mice. RF9 was found to have no effects at all on GnRH neurons in GnRH-GFP;Kiss1r-null mice, although these cells exhibited normal intrinsic electrical properties and remained responsive to GABA and glutamate. This study reveals that RF9 directly activates GnRH neurons in the mouse and that this is dependent upon Kiss1r expression.
Collapse
Affiliation(s)
- Xinhuai Liu
- Centre for Neuroendocrinology and Department of Physiology, University of Otago, School of Medical Sciences, Dunedin 9054, New Zealand
| | | |
Collapse
|
35
|
Evans MC, Rizwan M, Mayer C, Boehm U, Anderson GM. Evidence that insulin signalling in gonadotrophin-releasing hormone and kisspeptin neurones does not play an essential role in metabolic regulation of fertility in mice. J Neuroendocrinol 2014; 26:468-79. [PMID: 24824308 DOI: 10.1111/jne.12166] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 04/23/2014] [Accepted: 05/08/2014] [Indexed: 12/31/2022]
Abstract
Insulin in the brain plays an important role in regulating reproductive function, as demonstrated via conditional brain-specific insulin receptor (Insr) deletion (knockout). However, the specific neuronal target cells mediating the central effects of insulin on the reproductive axis remain unidentified. We first investigated whether insulin can act via direct effects on gonadotrophin-releasing hormone (GnRH) neurones. After clearly detecting Insr mRNA in an immunopurified GnRH cell fraction, we confirmed the presence of insulin receptor protein (InsR) in approximately 82% of GnRH neurones using dual-label immunohistochemistry. However, we did not observe any insulin-induced phospho-Akt (pAkt) or phospho-extracellular-signal-regulated kinase 1/2 in GnRH neurones, and therefore we investigated whether insulin signals via kisspeptin neurones to modulate GnRH release. Using dual-label immunohistochemistry, InsRs were detected only in approximately 5% of kisspeptin-immunoreactive cells. Insulin-induced pAkt was not observed in any kisspeptin-immunoreactive cells in either the rostral periventricular region of the third ventricle or arcuate nucleus in response to 200 mU of insulin treatment, although a more pharmacological dose (10 U) induced pronounced (> 20%) pAkt-kisspeptin coexpression in both regions. To confirm that insulin signalling via kisspeptin neurones does not critically modulate reproductive function, we generated kisspeptin-specific InsR knockout (KIRKO) mice and assessed multiple reproductive and metabolic parameters. No significant differences in puberty onset, oestrous cyclicity or reproductive competency were observed in the female or male KIRKO mice compared to their control littermates. However, significantly decreased fasting insulin (P < 0.05) and a nonsignificant trend towards reduced body weight were observed in male KIRKO mice. Thus, InsR signalling in kisspeptin cells is not critical for puberty onset or reproductive competency, although it may have a small metabolic effect in males.
Collapse
Affiliation(s)
- M C Evans
- Centre for Neuroendocrinology and Department of Anatomy, University of Otago School of Medical Sciences, Dunedin, New Zealand
| | | | | | | | | |
Collapse
|
36
|
Will RG, Hull EM, Dominguez JM. Influences of dopamine and glutamate in the medial preoptic area on male sexual behavior. Pharmacol Biochem Behav 2014; 121:115-23. [DOI: 10.1016/j.pbb.2014.02.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 01/13/2014] [Accepted: 02/05/2014] [Indexed: 11/25/2022]
|
37
|
Clarke IJ. Interface between metabolic balance and reproduction in ruminants: focus on the hypothalamus and pituitary. Horm Behav 2014; 66:15-40. [PMID: 24568750 DOI: 10.1016/j.yhbeh.2014.02.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 02/13/2014] [Accepted: 02/15/2014] [Indexed: 01/24/2023]
Abstract
This article is part of a Special Issue "Energy Balance". The interface between metabolic regulators and the reproductive system is reviewed with special reference to the sheep. Even though sheep are ruminants with particular metabolic characteristics, there is a broad consensus across species in the way that the reproductive system is influenced by metabolic state. An update on the neuroendocrinology of reproduction indicates the need to account for the way that kisspeptin provides major drive to gonadotropin releasing hormone (GnRH) neurons and also mediates the feedback effects of gonadal steroids. The way that kisspeptin function is influenced by appetite regulating peptides (ARP) is considered. Another newly recognised factor is gonadotropin inhibitory hormone (GnIH), which has a dual function in that it suppresses reproductive function whilst also acting as an orexigen. Our understanding of the regulation of food intake and energy expenditure has expanded exponentially in the last 3 decades and historical perspective is provided. The function of the regulatory factors and the hypothalamic cellular systems involved is reviewed with special reference to the sheep. Less is known of these systems in the cow, especially the dairy cow, in which a major fertility issue has emerged in parallel with selection for increased milk production. Other endocrine systems--the hypothalamo-pituitary-adrenal axis, the growth hormone (GH) axis and the thyroid hormones--are influenced by metabolic state and are relevant to the interface between metabolic function and reproduction. Special consideration is given to issues such as season and lactation, where the relationship between metabolic hormones and reproductive function is altered.
Collapse
Affiliation(s)
- Iain J Clarke
- Monash University, Department of Physiology, Wellington Road, Clayton 3168, Australia.
| |
Collapse
|
38
|
Jayasena CN, Comninos AN, Nijher GMK, Abbara A, De Silva A, Veldhuis JD, Ratnasabapathy R, Izzi-Engbeaya C, Lim A, Patel DA, Ghatei MA, Bloom SR, Dhillo WS. Twice-daily subcutaneous injection of kisspeptin-54 does not abolish menstrual cyclicity in healthy female volunteers. J Clin Endocrinol Metab 2013; 98:4464-74. [PMID: 24030945 PMCID: PMC4111853 DOI: 10.1210/jc.2013-1069] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Kisspeptin is a critical hypothalamic regulator of reproductive function. Chronic kisspeptin administration causes profound tachyphylaxis in male monkeys and in women with functional hypothalamic amenorrhea. The pharmacological effects of chronic kisspeptin exposure in healthy women with normal menstrual cycles have not been studied previously. AIM Our aim was to determine the effects of follicular-phase kisspeptin-54 treatment on menstrual cyclicity in healthy women. METHODS We performed a prospective, single-blinded, 1-way crossover study. Healthy women received twice-daily sc injections of kisspeptin (6.4 nmol/kg) or 0.9% saline during menstrual days 7-14 (n = 5 per treatment arm). Serial assessments of basal reproductive hormones, ultrasound parameters, LH pulsatility, and acute sensitivity to GnRH and kisspeptin-54 injection were performed. RESULTS Menstrual cyclicity persisted in all women after follicular-phase kisspeptin-54 treatment. Chronic exposure to kisspeptin-54 did not abolish acute stimulation of LH after injection of kisspeptin-54 or GnRH. In addition, kisspeptin-54 treatment was associated with a shorter mean length of the menstrual cycle (mean length of menstrual cycle was 28.6 ± 1.4 days with saline vs 26.8 ± 3.1 days with kisspeptin, P < .01), earlier onset of highest recorded serum LH (mean menstrual day of highest LH was 15.2 ± 1.3 with saline vs 13.0 ± 1.9 with kisspeptin, P < .05), and earlier onset of the luteal phase (mean menstrual day of progesterone increase was 18.0 ± 2.1 with saline vs 15.8 ± 0.9 with kisspeptin, P < .05). CONCLUSION Our data suggest that 1 week of exogenous kisspeptin-54 does not abolish menstrual cyclicity in healthy women. Further work is needed to determine whether kisspeptin could be used to treat certain anovulatory disorders.
Collapse
Affiliation(s)
- C N Jayasena
- Department of Investigative Medicine, Imperial College London, Sixth Floor, Commonwealth Building, Hammersmith Hospital, Du Cane Road, London W12 ONN, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Popa SM, Moriyama RM, Caligioni CS, Yang JJ, Cho CM, Concepcion TL, Oakley AE, Lee IH, Sanz E, Amieux PS, Caraty A, Palmiter RD, Navarro VM, Chan YM, Seminara SB, Clifton DK, Steiner RA. Redundancy in Kiss1 expression safeguards reproduction in the mouse. Endocrinology 2013; 154:2784-94. [PMID: 23736293 PMCID: PMC3713212 DOI: 10.1210/en.2013-1222] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Kisspeptin (Kiss1) signaling to GnRH neurons is widely acknowledged to be a prerequisite for puberty and reproduction. Animals lacking functional genes for either kisspeptin or its receptor exhibit low gonadotropin secretion and infertility. Paradoxically, a recent study reported that genetic ablation of nearly all Kiss1-expressing neurons (Kiss1 neurons) does not impair reproduction, arguing that neither Kiss1 neurons nor their products are essential for sexual maturation. We posited that only minute quantities of kisspeptin are sufficient to support reproduction. If this were the case, animals having dramatically reduced Kiss1 expression might retain fertility, testifying to the redundancy of Kiss1 neurons and their products. To test this hypothesis and to determine whether males and females differ in the required amount of kisspeptin needed for reproduction, we used a mouse (Kiss1-CreGFP) that has a severe reduction in Kiss1 expression. Mice that are heterozygous and homozygous for this allele (Kiss1(Cre/+) and Kiss1(Cre/Cre)) have ∼50% and 95% reductions in Kiss1 transcript, respectively. We found that although male Kiss1(Cre/Cre) mice sire normal-sized litters, female Kiss1(Cre/Cre) mice exhibit significantly impaired fertility and ovulation. These observations suggest that males require only 5% of normal Kiss1 expression to be reproductively competent, whereas females require higher levels for reproductive success.
Collapse
Affiliation(s)
- Simina M Popa
- The Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Colledge WH, Doran J, Mei H. Model systems for studying kisspeptin signalling: mice and cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 784:481-503. [PMID: 23550020 DOI: 10.1007/978-1-4614-6199-9_22] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Kisspeptins are a family of overlapping neuropeptides, encoded by the Kiss1 gene, that are required for activation and maintenance of the mammalian reproductive axis. Kisspeptins act within the hypothalamus to stimulate release of gonadotrophic releasing hormone and activation of the pituitary-gonadal axis. Robust model systems are required to dissect the regulatory mechanisms that control Kiss1 neuronal activity and to examine the molecular consequences of kisspeptin signalling. While studies in normal animals have been important in this, transgenic mice with targeted mutations affecting the kisspeptin signalling pathway have played a significant role in extending our understanding of kisspeptin physiology. Knock-out mice recapitulate the reproductive defects associated with mutations in humans and provide an experimentally tractable model system to interrogate regulatory feedback mechanisms. In addition, transgenic mice with cell-specific expression of modulator proteins such as the CRE recombinase or fluorescent reporter proteins such as GFP allow more sophisticated analyses such as cell or gene ablation or electrophysiological profiling. At a less complex level, immortalized cell lines have been useful for studying the role of kisspeptin in cell migration and metastasis and examining the intracellular signalling events associated with kisspeptin signalling.
Collapse
|
41
|
García-Galiano D, Pineda R, Roa J, Ruiz-Pino F, Sánchez-Garrido MA, Castellano JM, Aguilar E, Navarro VM, Pinilla L, Tena-Sempere M. Differential modulation of gonadotropin responses to kisspeptin by aminoacidergic, peptidergic, and nitric oxide neurotransmission. Am J Physiol Endocrinol Metab 2012; 303:E1252-63. [PMID: 23011064 DOI: 10.1152/ajpendo.00250.2012] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Kisspeptins (Kp), products of the Kiss1 gene, have emerged as essential elements in the control of GnRH neurons and gonadotropic secretion. However, despite considerable progress in the field, to date limited attention has been paid to elucidate the potential interactions of Kp with other neurotransmitters known to centrally regulate the gonadotropic axis. We characterize herein the impact of manipulations of key aminoacidergic (glutamate and GABA), peptidergic (NKB, Dyn, and MCH), and gaseous [nitric oxide (NO)] neurotransmission on gonadotropin responses to Kp-10 in male rats. Blockade of ionotropic glutamate receptors (of the NMDA and non-NMDA type) variably decreased LH responses to Kp-10, whereas activation of both ionotropic and metabotropic receptors, which enhanced LH and FSH release per se, failed to further increase gonadotropin responses to Kp-10. In fact, coactivation of metabotropic receptors attenuated LH and FSH responses to Kp-10. Selective activation of GABA(A) receptors decreased Kp-induced gonadotropin secretion, whereas their blockade elicited robust LH and FSH bursts and protracted responses to Kp-10 when combined with GABA(B) receptor inhibition. Blockade of Dyn signaling (at κ-opioid receptors) enhanced LH responses to Kp-10, whereas activation of Dyn and NKB signaling modestly reduced Kp-induced LH and FSH release. Finally, MCH decreased basal LH secretion and modestly reduced FSH responses to Kp-10, whereas LH responses to Kp-10 were protracted after inhibition of NO synthesis. In summary, we present herein evidence for the putative roles of glutamate, GABA, Dyn, NKB, MCH, and NO in modulating gonadotropic responses to Kp in male rats. Our pharmacological data will help to characterize the central interactions and putative hierarchy of key neuroendocrine pathways involved in the control of the gonadotropic axis.
Collapse
Affiliation(s)
- David García-Galiano
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Dungan Lemko HM, Elias CF. Kiss of the mutant mouse: how genetically altered mice advanced our understanding of kisspeptin's role in reproductive physiology. Endocrinology 2012; 153:5119-29. [PMID: 23011921 PMCID: PMC3473196 DOI: 10.1210/en.2012-1494] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The kisspeptin system has emerged as one of the most important circuits within the central network governing reproduction. Although kisspeptin physiology has been examined in many species, much of our understanding of this system has come from mice. Recently, the study of several innovative strains of genetically engineered mouse models has revealed intriguing and unexpected insights into the functions of kisspeptin signaling in the hypothalamus. Here, we review the advancements in our knowledge of the central kisspeptin system through the use of mutant mice.
Collapse
Affiliation(s)
- Heather M Dungan Lemko
- Department of Internal Medicine, Division of Hypothalamic Research, University of Texas Southwestern Medical Center, Dallas, 75390, USA.
| | | |
Collapse
|
43
|
Szymanski L, Bakker J. Aromatase knockout mice show normal steroid-induced activation of gonadotrophin-releasing hormone neurones and luteinising hormone surges with a reduced population of kisspeptin neurones in the rostral hypothalamus. J Neuroendocrinol 2012; 24:1222-33. [PMID: 22577852 DOI: 10.1111/j.1365-2826.2012.02334.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We recently reported that female aromatase knockout (ArKO) mice show deficits in sexual behaviour and a decreased population of kisspeptin-immunoreactive neurones in the rostral periventricular area of the third ventricle (RP3V), resurrecting the question of whether oestradiol actively contributes to female-typical sexual differentiation. To further address this question, we assessed the capacity of ArKO mice to generate a steroid-induced luteinising hormone (LH) surge. Adult, gonadectomised wild-type (WT) and ArKO mice were given silastic oestradiol implants s.c. and, 1 week later, received s.c. injections of either oestradiol benzoate (EB) followed by progesterone, EB alone, or no additional steroids to activate gonadotrophin-releasing hormone (GnRH) neurones and generate an LH surge. Treatment with EB and progesterone induced significant Fos/GnRH double-labelling and, consequently, an LH surge in female WT and in ArKO mice of both sexes but not in male WT mice. ArKO mice of both sexes had fewer cells expressing Kiss-1 mRNA in the RP3V compared to female WT mice but had more Kiss-1 mRNA-expressing cells compared to WT males, reflecting an incomplete sexual differentiation of this system. To determine the number of cells expressing kisspeptin, the same experimental design was repeated in Experiment 2 with the addition of groups of WT and ArKO mice that were given EB + progesterone and sacrificed 2 h before the expected LH surge. No differences were observed in the number of kisspeptin-immunoreactive cells 2 h before and at the time of the LH surge. The finding that ArKO mice of both sexes have a competent LH surge system suggests that oestradiol has predominantly defeminising actions on the GnRH/LH surge system in males and that the steroid-induced LH surge can occur in females even with a greatly reduced population of kisspeptin neurones in the RP3V.
Collapse
Affiliation(s)
- L Szymanski
- GIGA Neurosciences, University of Liège, Liège, Belgium
| | | |
Collapse
|
44
|
Pinilla L, Aguilar E, Dieguez C, Millar RP, Tena-Sempere M. Kisspeptins and Reproduction: Physiological Roles and Regulatory Mechanisms. Physiol Rev 2012; 92:1235-316. [DOI: 10.1152/physrev.00037.2010] [Citation(s) in RCA: 529] [Impact Index Per Article: 44.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Procreation is essential for survival of species. Not surprisingly, complex neuronal networks have evolved to mediate the diverse internal and external environmental inputs that regulate reproduction in vertebrates. Ultimately, these regulatory factors impinge, directly or indirectly, on a final common pathway, the neurons producing the gonadotropin-releasing hormone (GnRH), which stimulates pituitary gonadotropin secretion and thereby gonadal function. Compelling evidence, accumulated in the last few years, has revealed that kisspeptins, a family of neuropeptides encoded by the Kiss1 gene and produced mainly by neuronal clusters at discrete hypothalamic nuclei, are pivotal upstream regulators of GnRH neurons. As such, kisspeptins have emerged as important gatekeepers of key aspects of reproductive maturation and function, from sexual differentiation of the brain and puberty onset to adult regulation of gonadotropin secretion and the metabolic control of fertility. This review aims to provide a comprehensive account of the state-of-the-art in the field of kisspeptin physiology by covering in-depth the consensus knowledge on the major molecular features, biological effects, and mechanisms of action of kisspeptins in mammals and, to a lesser extent, in nonmammalian vertebrates. This review will also address unsolved and contentious issues to set the scene for future research challenges in the area. By doing so, we aim to endow the reader with a critical and updated view of the physiological roles and potential translational relevance of kisspeptins in the integral control of reproductive function.
Collapse
Affiliation(s)
- Leonor Pinilla
- Department of Cell Biology, Physiology and Immunology, University of Córdoba; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III; and Instituto Maimónides de Investigaciones Biomédicas, Córdoba, Spain; Department of Physiology, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain; and Centre for Integrative Physiology, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Enrique Aguilar
- Department of Cell Biology, Physiology and Immunology, University of Córdoba; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III; and Instituto Maimónides de Investigaciones Biomédicas, Córdoba, Spain; Department of Physiology, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain; and Centre for Integrative Physiology, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Carlos Dieguez
- Department of Cell Biology, Physiology and Immunology, University of Córdoba; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III; and Instituto Maimónides de Investigaciones Biomédicas, Córdoba, Spain; Department of Physiology, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain; and Centre for Integrative Physiology, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Robert P. Millar
- Department of Cell Biology, Physiology and Immunology, University of Córdoba; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III; and Instituto Maimónides de Investigaciones Biomédicas, Córdoba, Spain; Department of Physiology, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain; and Centre for Integrative Physiology, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Manuel Tena-Sempere
- Department of Cell Biology, Physiology and Immunology, University of Córdoba; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III; and Instituto Maimónides de Investigaciones Biomédicas, Córdoba, Spain; Department of Physiology, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain; and Centre for Integrative Physiology, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
45
|
Kisspeptin-GPR54 signaling in mouse NO-synthesizing neurons participates in the hypothalamic control of ovulation. J Neurosci 2012; 32:932-45. [PMID: 22262891 DOI: 10.1523/jneurosci.4765-11.2012] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Reproduction is controlled in the brain by a neural network that drives the secretion of gonadotropin-releasing hormone (GnRH). Various permissive homeostatic signals must be integrated to achieve ovulation in mammals. However, the neural events controlling the timely activation of GnRH neurons are not completely understood. Here we show that kisspeptin, a potent activator of GnRH neuronal activity, directly communicates with neurons that synthesize the gaseous transmitter nitric oxide (NO) in the preoptic region to coordinate the progression of the ovarian cycle. Using a transgenic Gpr54-null IRES-LacZ knock-in mouse model, we demonstrate that neurons containing neuronal NO synthase (nNOS), which are morphologically associated with kisspeptin fibers, express the kisspeptin receptor GPR54 in the preoptic region, but not in the tuberal region of the hypothalamus. The activation of kisspeptin signaling in preoptic neurons promotes the activation of nNOS through its phosphorylation on serine 1412 via the AKT pathway and mimics the positive feedback effects of estrogens. Finally, we show that while NO release restrains the reproductive axis at stages of the ovarian cycle during which estrogens exert their inhibitory feedback, it is required for the kisspeptin-dependent preovulatory activation of GnRH neurons. Thus, interactions between kisspeptin and nNOS neurons may play a central role in regulating the hypothalamic-pituitary-gonadal axis in vivo.
Collapse
|
46
|
García-Galiano D, van Ingen Schenau D, Leon S, Krajnc-Franken MAM, Manfredi-Lozano M, Romero-Ruiz A, Navarro VM, Gaytan F, van Noort PI, Pinilla L, Blomenröhr M, Tena-Sempere M. Kisspeptin signaling is indispensable for neurokinin B, but not glutamate, stimulation of gonadotropin secretion in mice. Endocrinology 2012; 153:316-28. [PMID: 22067321 DOI: 10.1210/en.2011-1260] [Citation(s) in RCA: 128] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Kisspeptins (Kp), products of the Kiss1 gene that act via Gpr54 to potently stimulate GnRH secretion, operate as mediators of other regulatory signals of the gonadotropic axis. Mouse models of Gpr54 and/or Kiss1 inactivation have been used to address the contribution of Kp in the central control of gonadotropin secretion; yet, phenotypic and hormonal differences have been detected among the transgenic lines available. We report here a series of neuroendocrine analyses in male mice of a novel Gpr54 knockout (KO) model, generated by heterozygous crossing of a loxP-Gpr54/Protamine-Cre double mutant line. Gpr54-null males showed severe hypogonadotropic hypogonadism but retained robust responsiveness to GnRH. Gonadotropic responses to the agonist of ionotropic glutamate receptors, N-methyl-d-aspartate, were attenuated, but persisted, in Gpr54-null mice. In contrast, LH secretion after activation of metabotropic glutamate receptors was totally preserved in the absence of Gpr54 signaling. Detectable, albeit reduced, LH responses were also observed in Gpr54 KO mice after intracerebroventricular administration of galanin-like peptide or RF9, putative antagonist of neuropeptide FF receptors for the mammalian ortholog of gonadotropin-inhibiting hormone. In contrast, the stimulatory effect of senktide, agonist of neurokinin B (NKB; cotransmitter of Kiss1 neurons), was totally abrogated in Gpr54 KO males. Lack of Kp signaling also eliminated feedback LH responses to testosterone withdrawal. However, residual but sustained increases of FSH were detected in gonadectomized Gpr54 KO males, in which testosterone replacement failed to fully suppress circulating FSH levels. In sum, our study provides novel evidence for the relative importance of Kp-dependent vs. -independent actions of several key regulators of GnRH secretion, such as glutamate, galanin-like peptide, and testosterone. In addition, our data document for the first time the indispensable role of Kp signaling in mediating the stimulatory effects of NKB on LH secretion, thus supporting the hypothesis that NKB actions on GnRH neurons are indirectly mediated via its ability to regulate Kiss1 neuronal output.
Collapse
Affiliation(s)
- David García-Galiano
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Avda. Menéndez Pidal s/n, 14004 Córdoba, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Navarro VM, Tena-Sempere M. Neuroendocrine control by kisspeptins: role in metabolic regulation of fertility. Nat Rev Endocrinol 2011; 8:40-53. [PMID: 21912400 DOI: 10.1038/nrendo.2011.147] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The neurohormonal control of reproduction involves a hierarchical network of central and peripheral signals in the hypothalamic-pituitary-gonadal (HPG) axis. Development and function of this neuroendocrine system is the result of a lifelong delicate balance between endogenous regulators and environmental cues, including nutritional and metabolic factors. Kisspeptins are the peptide products of KISS1, which operate via the G-protein-coupled receptor GPR54 (also known as Kiss1R). These peptides have emerged as essential upstream regulators of neurons secreting gonadotropin-releasing hormone (GnRH), the major hypothalamic node for the stimulatory control of the HPG axis. They are potent elicitors of gonadotropin secretion in various species and physiological settings. Moreover, Kiss1 neurons in the hypothalamus participate in crucial features of reproductive maturation and function, such as brain-level sex differentiation, puberty onset and the neuroendocrine regulation of gonadotropin secretion and ovulation. Cotransmitters of Kiss1 neurons, such as neurokinin B, with roles in controlling the HPG axis have been identified by genetic, neuroanatomical and physiological studies. In addition, a putative role has been proposed for Kiss1 neurons in transmitting metabolic information to GnRH neurons, although the precise mechanisms are as yet unclear. In this Review, we present the major reproductive features of kisspeptins, especially their interplay with neurokinin B and potential roles in the metabolic control of puberty and fertility, and suggest new avenues for research.
Collapse
Affiliation(s)
- Victor M Navarro
- Department of Cell Biology, Physiology and Immunology and CIBERobn, Faculty of Medicine, University of Córdoba, Avenida Menéndez Pidal s/n, 14004 Córdoba, Spain
| | | |
Collapse
|
48
|
Hankir MK, Parkinson JR, Bloom SR, Bell JD. The effects of glutamate receptor agonists and antagonists on mouse hypothalamic and hippocampal neuronal activity shown through manganese enhanced MRI. Neuroimage 2011; 59:968-78. [PMID: 21925279 DOI: 10.1016/j.neuroimage.2011.08.063] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Revised: 08/05/2011] [Accepted: 08/21/2011] [Indexed: 11/17/2022] Open
Abstract
Manganese enhanced MRI (MEMRI) is an imaging paradigm that can be used to assess neuronal activity in vivo. Here we investigate, through the use of MEMRI, the influence of receptor dynamics on neuronal activity in the hypothalamus and hippocampus focusing on the glutamate receptor signalling system. We demonstrate that intraperitoneal (i.p.) administration of monosodium glutamate (MSG) and the ionotropic glutamate receptor (iGluR) agonists NMDA and AMPA resulted in significantly increased signal intensity (SI) in the arcuate nucleus (ARC), the suprachiasmatic nucleus (SCN) and the CA3 region of the hippocampus of mice consistent with increased neuronal activity. Administration of the NMDA receptor antagonist MK-801 resulted in significantly decreased SI in the paraventricular nucleus (PVN) consistent with decreased neuronal activity. Co-administration of MSG and the AMPA receptor antagonist NBQX attenuated the increase in SI observed in the ARC from MSG alone, suggesting MEMRI may be applicable to the study of receptor dynamics in vivo. We also observed that administration of the various iGluR agonists and antagonists modulated SI in the lateral ventricle and that high dose MSG (300 mg) caused a hitherto unseen enhancement in SI in the entire cortical/subarachnoid region. In conclusion, MEMRI reveals changes in neuronal activity in response to iGluR agonists and antagonists in the CNS in vivo as well as revealing multifaceted effects beyond those attributable to neuronal activity alone.
Collapse
Affiliation(s)
- Mohammed K Hankir
- Metabolic and Molecular Imaging Group, MRC Clinical Sciences Centre, Imperial College London, 3rd Floor Cyclotron Building, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK.
| | | | | | | |
Collapse
|
49
|
Synergic effects of estradiol and progesterone on regulation of the hypothalamic neuronal nitric oxide synthase expression in ovariectomized mice. Brain Res 2011; 1404:1-9. [DOI: 10.1016/j.brainres.2011.06.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2011] [Revised: 06/03/2011] [Accepted: 06/04/2011] [Indexed: 11/20/2022]
|
50
|
Abstract
Gonadotropin-releasing hormone (GnRH) neurons represent the final common output of signals from the brain that regulates reproductive function. A wide range of environmental factors impact GnRH neuron activity including disease, stress, nutrition, and seasonal cues, as well as gonadal steroid hormones. The CNS response is thought to be mediated, at least in part, through intermediate signaling molecules that affect GnRH neuronal activity. In vitro, GnRH neuronal cell lines respond to a variety of ligands that activate the Jak (Janus-activated kinase)/STAT (signal transducers and activators of transcription) intracellular signaling pathway. To determine its biological function in reproduction, we used Cre (cAMP response element)/LoxP technology to generate GnRH neuron-specific Jak2 conditional knock-out (Jak2 G(-/-)) mice. GnRH mRNA levels were reduced in Jak2 G(-/-) mice when compared with controls, while the number of GnRH neurons was equivalent, indicating a reduction in GnRH gene expression. Secretion of GnRH is also reduced as basal serum luteinizing hormone (LH) levels were significantly lower in female Jak2 G(-/-) mice while the pituitary responded normally to exogenous GnRH. Preovulatory LH surge levels were blunted in Jak2 G(-/-) mice, which was correlated with reduced GnRH neuronal activation as assessed by c-Fos. However, the activation of GnRH neurons following release from estrogen-negative feedback is retained. Female Jak2 G(-/-) mice exhibited significantly delayed puberty and first estrus, abnormal estrous cyclicity, and impaired fertility. These results demonstrate an essential role for Jak2 signaling in GnRH neurons for normal reproductive development and fertility in female mice.
Collapse
|