1
|
Kovács A, Szabó E, László K, Kertes E, Zagorácz O, Mintál K, Tóth A, Gálosi R, Berta B, Lénárd L, Hormay E, László B, Zelena D, Tóth ZE. Brain RFamide Neuropeptides in Stress-Related Psychopathologies. Cells 2024; 13:1097. [PMID: 38994950 PMCID: PMC11240450 DOI: 10.3390/cells13131097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/21/2024] [Accepted: 06/22/2024] [Indexed: 07/13/2024] Open
Abstract
The RFamide peptide family is a group of proteins that share a common C-terminal arginine-phenylalanine-amide motif. To date, the family comprises five groups in mammals: neuropeptide FF, LPXRFamides/RFamide-related peptides, prolactin releasing peptide, QRFP, and kisspeptins. Different RFamide peptides have their own cognate receptors and are produced by different cell populations, although they all can also bind to neuropeptide FF receptors with different affinities. RFamide peptides function in the brain as neuropeptides regulating key aspects of homeostasis such as energy balance, reproduction, and cardiovascular function. Furthermore, they are involved in the organization of the stress response including modulation of pain. Considering the interaction between stress and various parameters of homeostasis, the role of RFamide peptides may be critical in the development of stress-related neuropathologies. This review will therefore focus on the role of RFamide peptides as possible key hubs in stress and stress-related psychopathologies. The neurotransmitter coexpression profile of RFamide-producing cells is also discussed, highlighting its potential functional significance. The development of novel pharmaceutical agents for the treatment of stress-related disorders is an ongoing need. Thus, the importance of RFamide research is underlined by the emergence of peptidergic and G-protein coupled receptor-based therapeutic targets in the pharmaceutical industry.
Collapse
Affiliation(s)
- Anita Kovács
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Evelin Szabó
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Kristóf László
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Erika Kertes
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Olga Zagorácz
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Kitti Mintál
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Attila Tóth
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Rita Gálosi
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Bea Berta
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - László Lénárd
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Edina Hormay
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Bettina László
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Dóra Zelena
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (A.K.); (E.S.); (K.L.); (E.K.); (O.Z.); (K.M.); (A.T.); (R.G.); (B.B.); (L.L.); (E.H.); (B.L.)
| | - Zsuzsanna E. Tóth
- Department of Anatomy, Histology and Embryology, Semmelweis University, H1094 Budapest, Hungary
| |
Collapse
|
2
|
Hu KL, Chen Z, Li X, Cai E, Yang H, Chen Y, Wang C, Ju L, Deng W, Mu L. Advances in clinical applications of kisspeptin-GnRH pathway in female reproduction. Reprod Biol Endocrinol 2022; 20:81. [PMID: 35606759 PMCID: PMC9125910 DOI: 10.1186/s12958-022-00953-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 04/30/2022] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Kisspeptin is the leading upstream regulator of pulsatile and surge Gonadotrophin-Releasing Hormone secretion (GnRH) in the hypothalamus, which acts as the key governor of the hypothalamic-pituitary-ovary axis. MAIN TEXT Exogenous kisspeptin or its receptor agonist can stimulate GnRH release and subsequent physiological gonadotropin secretion in humans. Based on the role of kisspeptin in the hypothalamus, a broad application of kisspeptin and its receptor agonist has been recently uncovered in humans, including central control of ovulation, oocyte maturation (particularly in women at a high risk of ovarian hyperstimulation syndrome), test for GnRH neuronal function, and gatekeepers of puberty onset. In addition, the kisspeptin analogs, such as TAK-448, showed promising agonistic activity in healthy women as well as in women with hypothalamic amenorrhoea or polycystic ovary syndrome. CONCLUSION More clinical trials should focus on the therapeutic effect of kisspeptin, its receptor agonist and antagonist in women with reproductive disorders, such as hypothalamic amenorrhoea, polycystic ovary syndrome, and endometriosis.
Collapse
Affiliation(s)
- Kai-Lun Hu
- Center for Reproductive Medicine, Peking University Third Hospital, No.49 Huayuan North Road, Haidian District, Beijing, People's Republic of China, 100191
- Zhejiang MedicalTech Therapeutics Company, No.665 Yumeng Road, Wenzhou, People's Republic of China, 325200
| | - Zimiao Chen
- Department of Endocrinology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China, 325000
| | - Xiaoxue Li
- Zhejiang MedicalTech Therapeutics Company, No.665 Yumeng Road, Wenzhou, People's Republic of China, 325200
| | - Enci Cai
- Department of Nutrition and Food Science, College of Food, Agricultural and Natural Resource Sciences, University of Minnesota, Twin Cities, Minneapolis, MN, 55455, USA
| | - Haiyan Yang
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China, 325000
| | - Yi Chen
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China, 325000
| | - Congying Wang
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China, 325000
| | - Liping Ju
- Zhejiang MedicalTech Therapeutics Company, No.665 Yumeng Road, Wenzhou, People's Republic of China, 325200
| | - Wenhai Deng
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China, 325006.
| | - Liangshan Mu
- Zhejiang MedicalTech Therapeutics Company, No.665 Yumeng Road, Wenzhou, People's Republic of China, 325200.
| |
Collapse
|
3
|
Regulation of stress response on the hypothalamic-pituitary-gonadal axis via gonadotropin-inhibitory hormone. Front Neuroendocrinol 2022; 64:100953. [PMID: 34757094 DOI: 10.1016/j.yfrne.2021.100953] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/16/2021] [Accepted: 10/24/2021] [Indexed: 11/21/2022]
Abstract
Under stressful condition, reproductive function is impaired due to the activation of various components of the hypothalamic-pituitaryadrenal (HPA) axis, which can suppress the activity of the hypothalamic-pituitary-gonadal (HPG) axis at multiple levels. A hypothalamic neuropeptide, gonadotropin-inhibitory hormone (GnIH) is a key negative regulator of reproduction that governs the HPG axis. Converging lines of evidence have suggested that different stress types and their duration, such as physical or psychological, and acute or chronic, can modulate the GnIH system. To clarify the sensitivity and reactivity of the GnIH system in response to stress, we summarize and critically review the available studies that investigated the effects of various stressors, such as restraint, nutritional/metabolic and social stress, on GnIH expression and/or its neuronal activity leading to altered HPG action. In this review, we focus on GnIH as the potential novel mediator responsible for stress-induced reproductive dysfunction.
Collapse
|
4
|
Nitric oxide resets kisspeptin-excited GnRH neurons via PIP2 replenishment. Proc Natl Acad Sci U S A 2021; 118:2012339118. [PMID: 33443156 DOI: 10.1073/pnas.2012339118] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Fertility relies upon pulsatile release of gonadotropin-releasing hormone (GnRH) that drives pulsatile luteinizing hormone secretion. Kisspeptin (KP) neurons in the arcuate nucleus are at the center of the GnRH pulse generation and the steroid feedback control of GnRH secretion. However, KP evokes a long-lasting response in GnRH neurons that is hard to reconcile with periodic GnRH activity required to drive GnRH pulses. Using calcium imaging, we show that 1) the tetrodotoxin-insensitive calcium response evoked by KP relies upon the ongoing activity of canonical transient receptor potential channels maintaining voltage-gated calcium channels in an activated state, 2) the duration of the calcium response is determined by the rate of resynthesis of phosphatidylinositol 4,5-bisphosphate (PIP2), and 3) nitric oxide terminates the calcium response by facilitating the resynthesis of PIP2 via the canonical pathway guanylyl cyclase/3',5'-cyclic guanosine monophosphate/protein kinase G. In addition, our data indicate that exposure to nitric oxide after KP facilitates the calcium response to a subsequent KP application. This effect was replicated using electrophysiology on GnRH neurons in acute brain slices. The interplay between KP and nitric oxide signaling provides a mechanism for modulation of the refractory period of GnRH neurons after KP exposure and places nitric oxide as an important component for tonic GnRH neuronal pulses.
Collapse
|
5
|
Constantin S, Pizano K, Matson K, Shan Y, Reynolds D, Wray S. An Inhibitory Circuit From Brainstem to GnRH Neurons in Male Mice: A New Role for the RFRP Receptor. Endocrinology 2021; 162:6132086. [PMID: 33564881 PMCID: PMC8016070 DOI: 10.1210/endocr/bqab030] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Indexed: 12/31/2022]
Abstract
RFamide-related peptides (RFRPs, mammalian orthologs of gonadotropin-inhibitory hormone) convey circadian, seasonal, and social cues to the reproductive system. They regulate gonadotropin secretion by modulating gonadotropin-releasing hormone (GnRH) neurons via the RFRP receptor. Mice lacking this receptor are fertile but exhibit abnormal gonadotropin responses during metabolic challenges, such as acute fasting, when the normal drop in gonadotropin levels is delayed. Although it is known that these food intake signals to the reproductive circuit originate in the nucleus tractus solitarius (NTS) in the brainstem, the phenotype of the neurons conveying the signal remains unknown. Given that neuropeptide FF (NPFF), another RFamide peptide, resides in the NTS and can bind to the RFRP receptor, we hypothesized that NPFF may regulate GnRH neurons. To address this question, we used a combination of techniques: cell-attached electrophysiology on GnRH-driven green fluorescent protein-tagged neurons in acute brain slices; calcium imaging on cultured GnRH neurons; and immunostaining on adult brain tissue. We found (1) NPFF inhibits GnRH neuron excitability via the RFRP receptor and its canonical signaling pathway (Gi/o protein and G protein-coupled inwardly rectifying potassium channels), (2) NPFF-like fibers in the vicinity of GnRH neurons coexpress neuropeptide Y, (3) the majority of NPFF-like cell bodies in the NTS also coexpress neuropeptide Y, and (4) acute fasting increased NPFF-like immunoreactivity in the NTS. Together these data indicate that NPFF neurons within the NTS inhibit GnRH neurons, and thus reproduction, during fasting but prior to the energy deficit.
Collapse
Affiliation(s)
- Stephanie Constantin
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke/National Institutes of Health, Bethesda, MD 20892-3703, USA
| | - Katherine Pizano
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke/National Institutes of Health, Bethesda, MD 20892-3703, USA
| | - Kaya Matson
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke/National Institutes of Health, Bethesda, MD 20892-3703, USA
| | - Yufei Shan
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke/National Institutes of Health, Bethesda, MD 20892-3703, USA
| | - Daniel Reynolds
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke/National Institutes of Health, Bethesda, MD 20892-3703, USA
| | - Susan Wray
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke/National Institutes of Health, Bethesda, MD 20892-3703, USA
- Correspondence: Dr. Susan Wray, Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 35 Convent Drive MSC 3703, Building 35, Room 3A1012, Bethesda, MD 20892, USA.
| |
Collapse
|
6
|
Ullah R, Naz R, Batool A, Wazir M, Rahman TU, Nabi G, Wahab F, Fu J, Shahab M. RF9 Rescues Cortisol-Induced Repression of Testosterone Levels in Adult Male Macaques. Front Physiol 2021; 12:630796. [PMID: 33716777 PMCID: PMC7946976 DOI: 10.3389/fphys.2021.630796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 02/05/2021] [Indexed: 12/03/2022] Open
Abstract
Cortisol inhibits hypothalamic-pituitary-gonadal (HPG) axis whereas RF9, a potent agonist of kisspeptin receptor (GPR54) activates HPG-axis during fasting-induced stress and under normal physiological conditions. However, the effect of RF9 on the cortisol-induced repressed HPG-axis is not studied yet. This study investigated whether exogenous cortisol-induced repression of the HPG-axis can be rescued by RF9. Six intact adult male rhesus monkeys (Macaca mulatta) habituated to chair-restraint were administered hydrocortisone sodium succinate at a rate of 20 mg/kg of body weight (BW) per day for 12 days. Single blood sample was taken by venipuncture from each animal on alternate days for hormones analyses. On experimental day 12, hydrocortisone treated monkeys received a single intravenous bolus of RF9 (n = 3) and vehicle (n = 3). The animals were bled for a period of 4 h at 60 min intervals from an indwelling cannula in the saphenous vein. RF9 was administered intravenously at the dose of 0.1 mg/kg BW immediately after taking 0 min sample. Plasma cortisol and testosterone concentrations were measured by using specific enzyme immunoassays. Hydrocortisone treatment increased plasma cortisol levels (P ≤ 0.0001) and decreased plasma testosterone (P ≤ 0.0127) levels. Interestingly, compared to vehicle, RF9 treatment significantly increased plasma testosterone levels at 120 min (P ≤ 0.0037), 180 min (P ≤ 0.0016), and 240 min (P ≤ 0.0001) intervals in the hydrocortisone treated monkeys. From these results, we concluded that RF9 administration relieves the suppressed HPG-axis in term of plasma testosterone levels in the cortisol treated monkeys.
Collapse
Affiliation(s)
- Rahim Ullah
- Department of Endocrinology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Rabia Naz
- Laboratory of Reproductive Neuroendocrinology, Department of Animal Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Aalia Batool
- Laboratory of Reproductive Neuroendocrinology, Department of Animal Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Madiha Wazir
- Laboratory of Reproductive Neuroendocrinology, Department of Animal Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Tanzil Ur Rahman
- Laboratory of Reproductive Neuroendocrinology, Department of Animal Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Ghulam Nabi
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Science, Hebei Normal University, Shijiazhuang, China
| | - Fazal Wahab
- Department of Biomedical Sciences, Pak-Austria Fachhochschule: Institute of Applied Sciences and Technology, Haripur, Pakistan
| | - Junfen Fu
- Department of Endocrinology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Muhammad Shahab
- Laboratory of Reproductive Neuroendocrinology, Department of Animal Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| |
Collapse
|
7
|
Nguyen T, Marusich J, Li JX, Zhang Y. Neuropeptide FF and Its Receptors: Therapeutic Applications and Ligand Development. J Med Chem 2020; 63:12387-12402. [PMID: 32673481 DOI: 10.1021/acs.jmedchem.0c00643] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The endogenous neuropeptide FF (NPFF) and its two cognate G protein-coupled receptors, Neuropeptide FF Receptors 1 and 2 (NPFFR1 and NPFFR2), represent a relatively new target system for many therapeutic applications including pain regulation, modulation of opioid side effects, drug reward, anxiety, cardiovascular conditions, and other peripheral effects. Since the cloning of NPFFR1 and NPFFR2 in 2000, significant progress has been made to understand their pharmacological roles and interactions with other receptor systems, notably the opioid receptors. A variety of NPFFR ligands with different mechanisms of action (agonists or antagonists) have been discovered although with limited subtype selectivities. Differential pharmacological effects have been observed for many of these NPFFR ligands, depending on assays/models employed and routes of administration. In this Perspective, we highlight the therapeutic potentials, current knowledge gaps, and latest updates of the development of peptidic and small molecule NPFFR ligands as tool compounds and therapeutic candidates.
Collapse
Affiliation(s)
- Thuy Nguyen
- Center for Drug Discovery, Research Triangle Institute, 3040 East Cornwallis Road, Research Triangle Park, North Carolina 27709, United States
| | - Julie Marusich
- Center for Drug Discovery, Research Triangle Institute, 3040 East Cornwallis Road, Research Triangle Park, North Carolina 27709, United States
| | - Jun-Xu Li
- Department of Pharmacology and Toxicology, University at Buffalo, the State University of New York, Buffalo, New York 14203, United States
| | - Yanan Zhang
- Center for Drug Discovery, Research Triangle Institute, 3040 East Cornwallis Road, Research Triangle Park, North Carolina 27709, United States
| |
Collapse
|
8
|
Gulcu Bulmus F, Canpolat S, Sahin Z, Bulmus O, Serhatlioglu I, Kelestimur H. Kisspeptin and RF9 prevent paroxetine-induced changes in some parameters of seminal vesicle fluid in the male rats. Andrologia 2020; 52:e13538. [PMID: 32052480 DOI: 10.1111/and.13538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 12/30/2019] [Accepted: 01/11/2020] [Indexed: 11/28/2022] Open
Abstract
The aim of the study was to examine possible impacts of paroxetine and agomelatine on the levels of some components that constitute the seminal vesicle fluid. As a second purpose, it was also aimed to examine how possible negative effects induced by paroxetine on seminal vesicle fluid components were affected by kisspeptin and RF9 (an RFamide-related peptide antagonist, RFRP). Forty-two male rats, aged 21 days, divided into six groups; control, sham, paroxetine, agomelatine, paroxetine + kisspeptin and paroxetine + RF9. Paroxetine (3.6 mg/kg) and agomelatine (10 mg/kg) were administrated by oral gavage. Kisspeptin (1 nmol) and RF9 (20 nmol) were administered intracerebroventricular (i.c.v). The experiments were ended on post-natal 120 days; fructose, vitamin E, sodium, potassium and magnesium levels were measured in seminal vesicle fluid. Fructose, vitamin E, magnesium and potassium levels were significantly decreased in seminal vesicle fluid from the rats treated with paroxetine but did not show significant differences following agomelatine administration. The co-administration of kisspeptin or RF9 with paroxetine prevented the paroxetine-induced negative effects on seminal vesicle fluid components. These results suggest that reduction in sperm fertilising ability caused by changes in seminal vesicle fluid can be seen in long-term antidepressant use. RF-9 and kisspeptin might have positive effects on long-term antidepressant use-induced infertility.
Collapse
Affiliation(s)
| | - Sinan Canpolat
- Department of Physiology, Medicine Faculty, Firat University, Elazig, Turkey
| | - Zafer Sahin
- Department of Physiology, Medicine Faculty, Karadeniz Technical University, Trabzon, Turkey
| | - Ozgur Bulmus
- Department of Physical Therapy and Rehabilitation, Faculty of Health Sciences, Firat University, Elazig, Turkey
| | - Ihsan Serhatlioglu
- Department of Biophysics, Medicine Faculty, Firat University, Elazig, Turkey
| | - Haluk Kelestimur
- Department of Physiology, Medicine Faculty, Firat University, Elazig, Turkey
| |
Collapse
|
9
|
Sahin Z, Kelestimur H. RF9: May it be a new therapeutic option for hypogonadotropic hypogonadism? Med Hypotheses 2019; 128:54-57. [PMID: 31203909 DOI: 10.1016/j.mehy.2019.05.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 04/30/2019] [Accepted: 05/12/2019] [Indexed: 12/12/2022]
Abstract
Hypogonadotropic hypogonadism (secondary hypogonadism), congenital or acquired, is a form of hypogonadism that is due to problems with either the hypothalamus or pituitary gland affecting gonadotropin levels. Pulsatile secretion of gonadotropin-releasing hormone (GnRH) by hypothalamus is a primer step to initiate the release of pituitary gonadotropins. Kisspeptin and gonadotropin-inhibitory hormone (GnIH) are accepted as two major players in the activation and inhibition of GnRH regarding the neuroendocrine functioning of the hypothalamic pituitary gonadal axis. Kisspeptin is known as the most potent activator of GnRH. Regarding the inhibition of GnRH, RF-amide-related peptide-3 (RFRP-3) is accepted as the mammalian orthologue of GnIH in avian species. RF9 (1-adamantane carbonyl-Arg-Phe-NH2) is an antagonist of RFRP-3/GnIH receptor (neuropeptide FF receptor 1 (NPFFR1; also termed as GPR147). In recent years, several studies have indicated that RF9 activates GnRH neurons and gonadotropins in a kisspeptin receptor (Kiss1r, formerly known as GPR54) dependent manner. These results suggest that RF9 may have a bimodal function as both an RFRP-3 antagonist and a kisspeptin agonist or it may be a kiss1r agonist rather than an RFRP-3/GnIH receptor antagonist. These interactions are possible because Kisspeptin and GnIH are members of the RF-amide family, and both possibilities are not far from explaining the potent gonadotropin stimulating effects of RF9. Therefore, we hypothesize that RF9 may be a new therapeutic option for the hypogonadotropic hypogonadism due to its potent GnRH stimulating effects. A constant or repeated administration of RF9 provides a sustained increase in plasma gonadotrophin levels. However, applications in the same way with GnRH analogues and kisspeptin may result in desensitization of the gonadotropic axis. The reasons reported above contribute to our hypothesis that RF9 may be a good option in the GnRH stimulating as a kisspeptin agonist. We suggest that further studies are needed to elucidate the potential effects of RF9 in the treatment of the hypogonadotropic hypogonadism.
Collapse
Affiliation(s)
- Zafer Sahin
- Department of Physiology, Karadeniz Technical University, Trabzon, Turkey.
| | | |
Collapse
|
10
|
Hu KL, Chang HM, Li R, Yu Y, Qiao J. Regulation of LH secretion by RFRP-3 - From the hypothalamus to the pituitary. Front Neuroendocrinol 2019; 52:12-21. [PMID: 29608929 DOI: 10.1016/j.yfrne.2018.03.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 02/12/2018] [Accepted: 03/29/2018] [Indexed: 12/17/2022]
Abstract
RFamide-related peptides (RFRPs) have long been identified as inhibitors of the hypothalamus-pituitary-gonad axis in mammals. However, less progress has been made in the detailed roles of RFRPs in the control of LH secretion. Recent studies have suggested that RFRP-3 neurons in the hypothalamus can regulate the secretion of LH at different levels, including kisspeptin neurons, GnRH neurons, and the pituitary. Additionally, conflicting results regarding the effects of RFRP-3 on these levels exist. In this review, we collect the latest evidence related to the effects of RFRP-3 neurons in regulating LH secretion by acting on kisspeptin neurons, GnRH neurons, and the pituitary and discuss the potential role of the timely reduction of RFRP-3 signaling in the modulation of the preovulatory LH surge.
Collapse
Affiliation(s)
- Kai-Lun Hu
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
| | - Hsun-Ming Chang
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; Department of Obstetrics and Gynaecology, University of British Columbia, British Columbia Children's Hospital Research Institute, Vancouver, British Columbia V5Z 4H4, Canada
| | - Rong Li
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
| | - Yang Yu
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China.
| | - Jie Qiao
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
11
|
Wolfe A, Hussain MA. The Emerging Role(s) for Kisspeptin in Metabolism in Mammals. Front Endocrinol (Lausanne) 2018; 9:184. [PMID: 29740399 PMCID: PMC5928256 DOI: 10.3389/fendo.2018.00184] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 04/05/2018] [Indexed: 12/17/2022] Open
Abstract
Kisspeptin was initially identified as a metastasis suppressor. Shortly after the initial discovery, a key physiologic role for kisspeptin emerged in the regulation of fertility, with kisspeptin acting as a neurotransmitter via the kisspeptin receptor, its cognate receptor, to regulate hypothalamic GnRH neurons, thereby affecting pituitary-gonadal function. Recent work has demonstrated a more expansive role for kisspeptin signaling in a variety of organ systems. Kisspeptin has been revealed as a significant player in regulating glucose homeostasis, feeding behavior, body composition as well as cardiac function. The direct impact of kisspeptin on peripheral metabolic tissues has only recently been recognized. Here, we review the emerging endocrine role of kisspeptin in regulating metabolic function. Controversies and current limitations in the field as well as areas of future studies toward kisspeptin's diverse array of functions will be highlighted.
Collapse
Affiliation(s)
- Andrew Wolfe
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD, United States
| | - Mehboob A. Hussain
- Department of Internal Medicine Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, United States
| |
Collapse
|
12
|
Ullah R, Batool A, Wazir M, Naz R, Rahman TU, Wahab F, Shahab M, Fu J. Gonadotropin inhibitory hormone and RF9 stimulate hypothalamic-pituitary-adrenal axis in adult male rhesus monkeys. Neuropeptides 2017; 66:1-7. [PMID: 28757099 DOI: 10.1016/j.npep.2017.07.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 07/19/2017] [Accepted: 07/23/2017] [Indexed: 12/17/2022]
Abstract
Stress activates gonadotropin inhibitory hormone (GnIH), hypothalamic-pituitary-adrenal axis (HPA-axis) and represses hypothalamic-pituitary-gonadal axis (HPG-axis) but RF9 administration relieves stress-induced repression of the HPG-axis. Importantly, it was not known whether GnIH signaling and RF9 synthetic peptide modulate the HPA axis. To assess this, mammalian orthologs of GnIH (RFRP-1 and RFRP-3) and RF9 were administered to intact adult male rhesus monkeys. RFRP-1 (125μg/animal), RFRP-3 (250μg/animal) and RF9 (0.1mg/kg BW) were intravenously (iv) injected into normal fed (n=4) monkeys. Additionally, a single bolus iv injection of RF9 (0.1mg/kg BW) was also administered to 48h fasted monkeys (n=4) to check the effects of RF9 signaling on an activated HPA-axis. Serial blood samples were collected, centrifuged and the obtained plasma was used for the analysis of cortisol by specific enzyme immunoassay. RFRP-1 treatment significantly increased cortisol levels while RFRP-3 increased the plasma cortisol, but the effect was non-significant. RF9 treatment significantly increased cortisol levels in normal fed animals. In contrast, RF9 injection did not significantly alter circulating cortisol in fasted monkeys. In conclusion, our results suggest stimulatory action of RFRPs and RF9 on the HPA axis in the adult male monkeys. However, the mechanism and site of action of RFRP-1 and RF9 along the HPA-axis is still unknown. Therefore, further studies are needed to decipher the mechanism and site of action of RFRPs and RF9 on the HPA axis in primates.
Collapse
Affiliation(s)
- Rahim Ullah
- Department of Endocrinology, Children's Hospital of Zhejiang University School of Medicine, Hangzhou 310051, China; Laboratory of Reproductive Neuroendocrinology, Department of Animal Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Aalia Batool
- Laboratory of Reproductive Neuroendocrinology, Department of Animal Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan; The State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing 100101, China
| | - Madiha Wazir
- Laboratory of Reproductive Neuroendocrinology, Department of Animal Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Rabia Naz
- Laboratory of Reproductive Neuroendocrinology, Department of Animal Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Tanzil Ur Rahman
- Laboratory of Reproductive Neuroendocrinology, Department of Animal Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan; Department of Pathology and Pathophysiology, Key Laboratory of Reproductive Genetics, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Fazal Wahab
- Platform Degenerative Diseases, German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, D-37077 Göttingen, Germany
| | - Muhammad Shahab
- Laboratory of Reproductive Neuroendocrinology, Department of Animal Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| | - Junfen Fu
- Department of Endocrinology, Children's Hospital of Zhejiang University School of Medicine, Hangzhou 310051, China.
| |
Collapse
|
13
|
Korthanke CM, Thorson JF, Prezotto LD, Welsh TH, Cardoso RC, Williams GL. Secretion of Gonadotropins in Response to a Novel Kiss-1 Receptor Agonist, RF9 in the Mare: Modulation by Estradiol-17β and Half-Life of RF9 in the Peripheral Circulation. J Equine Vet Sci 2017. [DOI: 10.1016/j.jevs.2017.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
14
|
Elhabazi K, Humbert JP, Bertin I, Quillet R, Utard V, Schneider S, Schmitt M, Bourguignon JJ, Laboureyras E, Ben Boujema M, Simonnet G, Ancel C, Simonneaux V, Beltramo M, Bucher B, Sorg T, Meziane H, Schneider E, Petit-Demoulière B, Ilien B, Bihel F, Simonin F. RF313, an orally bioavailable neuropeptide FF receptor antagonist, opposes effects of RF-amide-related peptide-3 and opioid-induced hyperalgesia in rodents. Neuropharmacology 2017; 118:188-198. [DOI: 10.1016/j.neuropharm.2017.03.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 02/27/2017] [Accepted: 03/06/2017] [Indexed: 02/08/2023]
|
15
|
Thorson JF, Heidorn NL, Ryu V, Czaja K, Nonneman DJ, Barb CR, Hausman GJ, Rohrer GA, Prezotto LD, McCosh RB, Wright EC, White BR, Freking BA, Oliver WT, Hileman SM, Lents CA. Relationship of neuropeptide FF receptors with pubertal maturation of gilts †. Biol Reprod 2017; 96:617-634. [DOI: 10.1095/biolreprod.116.144998] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 01/01/2017] [Indexed: 01/14/2023] Open
|
16
|
Kim JS. What's in a Name? Roles of RFamide-Related Peptides Beyond Gonadotrophin Inhibition. J Neuroendocrinol 2016; 28. [PMID: 27369805 DOI: 10.1111/jne.12407] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 06/21/2016] [Accepted: 06/23/2016] [Indexed: 12/29/2022]
Abstract
RFamide-related peptides (RFRPs) have been heavily implicated in the control of reproductive function subsequent to their discovery more than 16 years ago. However, recent studies using genetic and pharmacological tools have challenged their importance in regulating the hypothalamic-pituitary-gonadal axis. It is generally accepted that RFRPs act as part of a wider RFamide system, which involves two receptors, called the neuropeptide FF receptors (NPFFR1 and R2), and includes the closely-related neuropeptide NPFF. NPFF has been studied ever since the 1980s and many of the functions of NPFF are also shared by RFRPs. The current review questions whether these functions of NPFF are indeed specific to just NPFF alone and presents evidence from both neuroendocrine and pharmacological perspectives. Furthermore, recently emerging new functions of RFRPs are discussed with the overall goal of clarifying the functions of RFRPs beyond the hypothalamic-pituitary-gonadal axis.
Collapse
Affiliation(s)
- J S Kim
- Centre for Neuroendocrinology and Department of Anatomy, University of Otago, Dunedin, New Zealand
| |
Collapse
|
17
|
Dardente H, Lomet D, Robert V, Decourt C, Beltramo M, Pellicer-Rubio MT. Seasonal breeding in mammals: From basic science to applications and back. Theriogenology 2016; 86:324-32. [DOI: 10.1016/j.theriogenology.2016.04.045] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 02/02/2016] [Accepted: 03/14/2016] [Indexed: 12/29/2022]
|
18
|
Liu X, Herbison AE. Kisspeptin Regulation of Neuronal Activity throughout the Central Nervous System. Endocrinol Metab (Seoul) 2016; 31:193-205. [PMID: 27246282 PMCID: PMC4923402 DOI: 10.3803/enm.2016.31.2.193] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 04/14/2016] [Accepted: 04/22/2016] [Indexed: 12/30/2022] Open
Abstract
Kisspeptin signaling at the gonadotropin-releasing hormone (GnRH) neuron is now relatively well characterized and established as being critical for the neural control of fertility. However, kisspeptin fibers and the kisspeptin receptor (KISS1R) are detected throughout the brain suggesting that kisspeptin is involved in regulating the activity of multiple neuronal circuits. We provide here a review of kisspeptin actions on neuronal populations throughout the brain including the magnocellular oxytocin and vasopressin neurons, and cells within the arcuate nucleus, hippocampus, and amygdala. The actions of kisspeptin in these brain regions are compared to its effects upon GnRH neurons. Two major themes arise from this analysis. First, it is apparent that kisspeptin signaling through KISS1R at the GnRH neuron is a unique, extremely potent form or neurotransmission whereas kisspeptin actions through KISS1R in other brain regions exhibit neuromodulatory actions typical of other neuropeptides. Second, it is becoming increasingly likely that kisspeptin acts as a neuromodulator not only through KISS1R but also through other RFamide receptors such as the neuropeptide FF receptors (NPFFRs). We suggest likely locations of kisspeptin signaling through NPFFRs but note that only limited tools are presently available for examining kisspeptin cross-signaling within the RFamide family of neuropeptides.
Collapse
Affiliation(s)
- Xinhuai Liu
- Centre for Neuroendocrinology, Department of Physiology, University of Otago School of Medical Sciences, Dunedin, New Zealand
| | - Allan E Herbison
- Centre for Neuroendocrinology, Department of Physiology, University of Otago School of Medical Sciences, Dunedin, New Zealand.
| |
Collapse
|
19
|
A synthetic kisspeptin analog that triggers ovulation and advances puberty. Sci Rep 2016; 6:26908. [PMID: 27245315 PMCID: PMC4887910 DOI: 10.1038/srep26908] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 05/09/2016] [Indexed: 12/26/2022] Open
Abstract
The neuropeptide kisspeptin and its receptor, KiSS1R, govern the reproductive timeline of mammals by triggering puberty onset and promoting ovulation by stimulating gonadotrophin-releasing hormone (GnRH) secretion. To overcome the drawback of kisspeptin short half-life we designed kisspeptin analogs combining original modifications, triazole peptidomimetic and albumin binding motif, to reduce proteolytic degradation and to slow down renal clearance, respectively. These analogs showed improved in vitro potency and dramatically enhanced pharmacodynamics. When injected intramuscularly into ewes (15 nmol/ewe) primed with a progestogen, the best analog (compound 6, C6) induced synchronized ovulations in both breeding and non-breeding seasons. Ovulations were fertile as demonstrated by the delivery of lambs at term. C6 was also fully active in both female and male mice but was completely inactive in KiSS1R KO mice. Electrophysiological recordings of GnRH neurons from brain slices of GnRH-GFP mice indicated that C6 exerted a direct excitatory action on GnRH neurons. Finally, in prepubertal female mice daily injections (0.3 nmol/mouse) for five days significantly advanced puberty. C6 ability to trigger ovulation and advance puberty demonstrates that kisspeptin analogs may find application in the management of livestock reproduction and opens new possibilities for the treatment of reproductive disorders in humans.
Collapse
|
20
|
Decourt C, Anger K, Robert V, Lomet D, Bartzen-Sprauer J, Caraty A, Dufourny L, Anderson G, Beltramo M. No Evidence That RFamide-Related Peptide 3 Directly Modulates LH Secretion in the Ewe. Endocrinology 2016; 157:1566-75. [PMID: 26862995 DOI: 10.1210/en.2015-1854] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The neuropeptide RFamide-related peptide 3 (RFRP-3) has been implicated in the control of gonadotropin secretion in both birds and mammals. However, in mammals, depending on species, sex and photoperiod, inhibitory, excitatory, or no effect of RFRP-3 on the plasma concentration of LH has been reported. In the ewe, treatment with RFRP-3 either reduced LH concentration or had no effect, and treatment with an RFRP-3 receptor antagonist (ie, RF9) resulted in increased concentration of plasma LH. To clarify these conflicting results in the present study, a set of experiments was performed in ewes. Multiple iv injections of RFRP-3 (6 × 50 μg) in ovariectomized ewes had no effect on plasma LH pulsatility. In intact ewes a bolus injection (500 μg) or an injection (250, 500, or 1000 μg) followed by a 4-hour perfusion (250, 500, or 1000 μg · h(-1)) of RFRP-3 had no effect on the LH pulse induced by kisspeptin (6.5 μg). In ovariectomized, estrogen-replaced ewes, the LH surge induced by estradiol benzoate was not modified by a 24-hour perfusion of RFRP-3 (500 μg h(-1)). Finally, although treatment with RF9 induced a robust release of LH, treatment with a more selective RFRP-3 receptor antagonist, GJ14, resulted in no evident increase of LH. In contrast to the inhibitory effect previously suggested, our data are more consistent with the concept that RFRP-3 has no direct effect on LH secretion in ewes and that RF9 effect on LH release is likely not RFRP-3 receptor mediated. Hence, RFRP-3 probably has a minor role on the control of LH secretion in the ewe.
Collapse
Affiliation(s)
- C Decourt
- Unité Mixte de Recherche Physiologie de la Reproduction et des Comportements (Institut National de la Recherche Agronomique, Unité Mixte de Recherche 85; Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7247, Université François Rabelais Tours, IFCE) (C.D., K.A., V.R., D.L., J.B.-S., A.C., L.D., M.B.), F-37380 Nouzilly, France; and Centre for Neuroendocrinology and Department of Anatomy (G.A.), University of Otago School of Medical Sciences, Dunedin 9054, New Zealand
| | - K Anger
- Unité Mixte de Recherche Physiologie de la Reproduction et des Comportements (Institut National de la Recherche Agronomique, Unité Mixte de Recherche 85; Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7247, Université François Rabelais Tours, IFCE) (C.D., K.A., V.R., D.L., J.B.-S., A.C., L.D., M.B.), F-37380 Nouzilly, France; and Centre for Neuroendocrinology and Department of Anatomy (G.A.), University of Otago School of Medical Sciences, Dunedin 9054, New Zealand
| | - V Robert
- Unité Mixte de Recherche Physiologie de la Reproduction et des Comportements (Institut National de la Recherche Agronomique, Unité Mixte de Recherche 85; Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7247, Université François Rabelais Tours, IFCE) (C.D., K.A., V.R., D.L., J.B.-S., A.C., L.D., M.B.), F-37380 Nouzilly, France; and Centre for Neuroendocrinology and Department of Anatomy (G.A.), University of Otago School of Medical Sciences, Dunedin 9054, New Zealand
| | - D Lomet
- Unité Mixte de Recherche Physiologie de la Reproduction et des Comportements (Institut National de la Recherche Agronomique, Unité Mixte de Recherche 85; Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7247, Université François Rabelais Tours, IFCE) (C.D., K.A., V.R., D.L., J.B.-S., A.C., L.D., M.B.), F-37380 Nouzilly, France; and Centre for Neuroendocrinology and Department of Anatomy (G.A.), University of Otago School of Medical Sciences, Dunedin 9054, New Zealand
| | - J Bartzen-Sprauer
- Unité Mixte de Recherche Physiologie de la Reproduction et des Comportements (Institut National de la Recherche Agronomique, Unité Mixte de Recherche 85; Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7247, Université François Rabelais Tours, IFCE) (C.D., K.A., V.R., D.L., J.B.-S., A.C., L.D., M.B.), F-37380 Nouzilly, France; and Centre for Neuroendocrinology and Department of Anatomy (G.A.), University of Otago School of Medical Sciences, Dunedin 9054, New Zealand
| | - A Caraty
- Unité Mixte de Recherche Physiologie de la Reproduction et des Comportements (Institut National de la Recherche Agronomique, Unité Mixte de Recherche 85; Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7247, Université François Rabelais Tours, IFCE) (C.D., K.A., V.R., D.L., J.B.-S., A.C., L.D., M.B.), F-37380 Nouzilly, France; and Centre for Neuroendocrinology and Department of Anatomy (G.A.), University of Otago School of Medical Sciences, Dunedin 9054, New Zealand
| | - L Dufourny
- Unité Mixte de Recherche Physiologie de la Reproduction et des Comportements (Institut National de la Recherche Agronomique, Unité Mixte de Recherche 85; Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7247, Université François Rabelais Tours, IFCE) (C.D., K.A., V.R., D.L., J.B.-S., A.C., L.D., M.B.), F-37380 Nouzilly, France; and Centre for Neuroendocrinology and Department of Anatomy (G.A.), University of Otago School of Medical Sciences, Dunedin 9054, New Zealand
| | - G Anderson
- Unité Mixte de Recherche Physiologie de la Reproduction et des Comportements (Institut National de la Recherche Agronomique, Unité Mixte de Recherche 85; Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7247, Université François Rabelais Tours, IFCE) (C.D., K.A., V.R., D.L., J.B.-S., A.C., L.D., M.B.), F-37380 Nouzilly, France; and Centre for Neuroendocrinology and Department of Anatomy (G.A.), University of Otago School of Medical Sciences, Dunedin 9054, New Zealand
| | - M Beltramo
- Unité Mixte de Recherche Physiologie de la Reproduction et des Comportements (Institut National de la Recherche Agronomique, Unité Mixte de Recherche 85; Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7247, Université François Rabelais Tours, IFCE) (C.D., K.A., V.R., D.L., J.B.-S., A.C., L.D., M.B.), F-37380 Nouzilly, France; and Centre for Neuroendocrinology and Department of Anatomy (G.A.), University of Otago School of Medical Sciences, Dunedin 9054, New Zealand
| |
Collapse
|
21
|
Quillet R, Ayachi S, Bihel F, Elhabazi K, Ilien B, Simonin F. RF-amide neuropeptides and their receptors in Mammals: Pharmacological properties, drug development and main physiological functions. Pharmacol Ther 2016; 160:84-132. [PMID: 26896564 DOI: 10.1016/j.pharmthera.2016.02.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
RF-amide neuropeptides, with their typical Arg-Phe-NH2 signature at their carboxyl C-termini, belong to a lineage of peptides that spans almost the entire life tree. Throughout evolution, RF-amide peptides and their receptors preserved fundamental roles in reproduction and feeding, both in Vertebrates and Invertebrates. The scope of this review is to summarize the current knowledge on the RF-amide systems in Mammals from historical aspects to therapeutic opportunities. Taking advantage of the most recent findings in the field, special focus will be given on molecular and pharmacological properties of RF-amide peptides and their receptors as well as on their implication in the control of different physiological functions including feeding, reproduction and pain. Recent progress on the development of drugs that target RF-amide receptors will also be addressed.
Collapse
Affiliation(s)
- Raphaëlle Quillet
- Biotechnologie et Signalisation Cellulaire, UMR 7242 CNRS, Université de Strasbourg, Illkirch, France
| | - Safia Ayachi
- Biotechnologie et Signalisation Cellulaire, UMR 7242 CNRS, Université de Strasbourg, Illkirch, France
| | - Frédéric Bihel
- Laboratoire Innovation Thérapeutique, UMR 7200 CNRS, Université de Strasbourg, Illkirch, France
| | - Khadija Elhabazi
- Biotechnologie et Signalisation Cellulaire, UMR 7242 CNRS, Université de Strasbourg, Illkirch, France
| | - Brigitte Ilien
- Biotechnologie et Signalisation Cellulaire, UMR 7242 CNRS, Université de Strasbourg, Illkirch, France
| | - Frédéric Simonin
- Biotechnologie et Signalisation Cellulaire, UMR 7242 CNRS, Université de Strasbourg, Illkirch, France.
| |
Collapse
|
22
|
Min L, Leon S, Li H, Pinilla L, Carroll RS, Tena-Sempere M, Kaiser UB. RF9 Acts as a KISS1R Agonist In Vivo and In Vitro. Endocrinology 2015; 156:4639-48. [PMID: 26418326 PMCID: PMC4655216 DOI: 10.1210/en.2015-1635] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
RF9, a reported antagonist of the mammalian gonadotropin-inhibitory hormone receptor, stimulates gonadotropin secretion in mammals. Recent studies have suggested that the stimulatory effect of RF9 on gonadotropin secretion relies on intact kisspeptin receptor (KISS1R) signaling, but the underlying mechanisms remain to be elucidated. Using Chinese Hamster Ovary cells stably transfected with KISS1R, we show that RF9 binds specifically to KISS1R, with a Kd of 1.6 × 10(-5)M, and stimulates an increase in intracellular calcium and inositol phosphate accumulation in a KISS1R-dependent manner, with EC50 values of 3.0 × 10(-6)M and 1.6 × 10(-7)M, respectively. RF9 also stimulated ERK phosphorylation, with a time course similar to that of kisspeptin-10. RFRP-3, the putative endogenous ligand for NPFFR1, did not stimulate inositol phosphate accumulation or pERK, nor did it alter responses to of kisspeptin-10 or RF9. In agreement with these in vitro data, we found that RF9 stimulated a robust LH increase in Npffr1(-/-) mice, similar to that in wild-type littermates, whereas the stimulatory effect of RF9 was markedly reduced in Kiss1r(-/-) and double Kiss1r(-/-)/Npfrr1(-/-) mice. The stimulatory effect of RF9 on LH secretion was restored by the selective rescue of Kiss1r expression in GnRH neurons, in Kiss1r(-/-T) mice. Taken together, our study demonstrates that RF9 acts primarily as a KISS1R agonist, but not as an allosteric modulator, to stimulate LH secretion. Our findings raise questions regarding the utility of RF9 for assessing NPFF1R function and de-emphasize a predominant role of this signaling system in central regulation of reproduction.
Collapse
Affiliation(s)
- Le Min
- Division of Endocrinology, Diabetes and Hypertension (L.M., H.L., R.S.C., U.B.K.), Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115; Department of Cell Biology, Physiology and Immunology (S.L., L.P., M.T.-S.), University of Córdoba; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, and Instituto Maimónides de Investigación Biomédica de Córdoba/Hospital Universitario Reina Sofia, Córdoba, 14004 Spain; and FiDiPro Program, Department of Physiology (M.T.-S.), University of Turku, FIN-20520 Turku, Finland
| | - Silvia Leon
- Division of Endocrinology, Diabetes and Hypertension (L.M., H.L., R.S.C., U.B.K.), Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115; Department of Cell Biology, Physiology and Immunology (S.L., L.P., M.T.-S.), University of Córdoba; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, and Instituto Maimónides de Investigación Biomédica de Córdoba/Hospital Universitario Reina Sofia, Córdoba, 14004 Spain; and FiDiPro Program, Department of Physiology (M.T.-S.), University of Turku, FIN-20520 Turku, Finland
| | - Huan Li
- Division of Endocrinology, Diabetes and Hypertension (L.M., H.L., R.S.C., U.B.K.), Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115; Department of Cell Biology, Physiology and Immunology (S.L., L.P., M.T.-S.), University of Córdoba; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, and Instituto Maimónides de Investigación Biomédica de Córdoba/Hospital Universitario Reina Sofia, Córdoba, 14004 Spain; and FiDiPro Program, Department of Physiology (M.T.-S.), University of Turku, FIN-20520 Turku, Finland
| | - Leonor Pinilla
- Division of Endocrinology, Diabetes and Hypertension (L.M., H.L., R.S.C., U.B.K.), Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115; Department of Cell Biology, Physiology and Immunology (S.L., L.P., M.T.-S.), University of Córdoba; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, and Instituto Maimónides de Investigación Biomédica de Córdoba/Hospital Universitario Reina Sofia, Córdoba, 14004 Spain; and FiDiPro Program, Department of Physiology (M.T.-S.), University of Turku, FIN-20520 Turku, Finland
| | - Rona S Carroll
- Division of Endocrinology, Diabetes and Hypertension (L.M., H.L., R.S.C., U.B.K.), Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115; Department of Cell Biology, Physiology and Immunology (S.L., L.P., M.T.-S.), University of Córdoba; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, and Instituto Maimónides de Investigación Biomédica de Córdoba/Hospital Universitario Reina Sofia, Córdoba, 14004 Spain; and FiDiPro Program, Department of Physiology (M.T.-S.), University of Turku, FIN-20520 Turku, Finland
| | - Manuel Tena-Sempere
- Division of Endocrinology, Diabetes and Hypertension (L.M., H.L., R.S.C., U.B.K.), Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115; Department of Cell Biology, Physiology and Immunology (S.L., L.P., M.T.-S.), University of Córdoba; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, and Instituto Maimónides de Investigación Biomédica de Córdoba/Hospital Universitario Reina Sofia, Córdoba, 14004 Spain; and FiDiPro Program, Department of Physiology (M.T.-S.), University of Turku, FIN-20520 Turku, Finland
| | - Ursula B Kaiser
- Division of Endocrinology, Diabetes and Hypertension (L.M., H.L., R.S.C., U.B.K.), Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115; Department of Cell Biology, Physiology and Immunology (S.L., L.P., M.T.-S.), University of Córdoba; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, and Instituto Maimónides de Investigación Biomédica de Córdoba/Hospital Universitario Reina Sofia, Córdoba, 14004 Spain; and FiDiPro Program, Department of Physiology (M.T.-S.), University of Turku, FIN-20520 Turku, Finland
| |
Collapse
|
23
|
Kim JS, Brownjohn PW, Dyer BS, Beltramo M, Walker CS, Hay DL, Painter GF, Tyndall JDA, Anderson GM. Anxiogenic and Stressor Effects of the Hypothalamic Neuropeptide RFRP-3 Are Overcome by the NPFFR Antagonist GJ14. Endocrinology 2015; 156:4152-62. [PMID: 26259035 DOI: 10.1210/en.2015-1532] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
RFamide-related peptide-3 (RFRP-3) is a recently discovered neuropeptide that has been proposed to play a role in the stress response. We aimed to elucidate the role of RFRP-3 and its receptor, neuropeptide FF (NPFF1R), in modulation of stress and anxiety responses. To achieve this, we characterized a new NPFF1R antagonist because our results showed that the only commercially available putative antagonist, RF9, is in fact an agonist at both NPFF1R and the kisspeptin receptor (KISS1R). We report here the identification and pharmacological characterization of GJ14, a true NPFFR antagonist. In in vivo tests of hypothalamic-pituitary-adrenal (HPA) axis function, GJ14 completely blocked RFRP-3-induced corticosterone release and neuronal activation in CRH neurons. Furthermore, chronic infusion of GJ14 led to anxiolytic-like behavior, whereas RFRP-3 infusion had anxiogenic effects. Mice receiving chronic RFRP-3 infusion also had higher basal circulating corticosterone levels. These results indicate a stimulatory action of RFRP-3 on the HPA axis, consistent with the dense expression of NPFF1R in the vicinity of CRH neurons. Importantly, coinfusion of RFRP-3 and GJ14 completely reversed the anxiogenic and HPA axis-stimulatory effects of RFRP-3. Here we have established the role of RFRP-3 as a regulator of stress and anxiety. We also show that GJ14 can reverse the effects of RFRP-3 both in vitro and in vivo. Infusion of GJ14 causes anxiolysis, revealing a novel potential target for treating anxiety disorders.
Collapse
Affiliation(s)
- Joon S Kim
- Centre for Neuroendocrinology and Department of Anatomy (J.S.K., P.W.B., G.M.A.) and National School of Pharmacy (J.D.A.T.), University of Otago, Dunedin 9054, New Zealand; Glycosyn (B.S.D.), Callaghan Innovation (B.S.D.), and The Ferrier Research Institute (G.F.P.), University of Wellington, Lower Hutt 5010, Victoria, New Zealand; Institut National de la Recherche Agronomique (M.B.), Nouzilly 37380, France; and School of Biological Sciences (C.S.W., D.L.H.), University of Auckland, Auckland 1142, New Zealand
| | - Phil W Brownjohn
- Centre for Neuroendocrinology and Department of Anatomy (J.S.K., P.W.B., G.M.A.) and National School of Pharmacy (J.D.A.T.), University of Otago, Dunedin 9054, New Zealand; Glycosyn (B.S.D.), Callaghan Innovation (B.S.D.), and The Ferrier Research Institute (G.F.P.), University of Wellington, Lower Hutt 5010, Victoria, New Zealand; Institut National de la Recherche Agronomique (M.B.), Nouzilly 37380, France; and School of Biological Sciences (C.S.W., D.L.H.), University of Auckland, Auckland 1142, New Zealand
| | - Blake S Dyer
- Centre for Neuroendocrinology and Department of Anatomy (J.S.K., P.W.B., G.M.A.) and National School of Pharmacy (J.D.A.T.), University of Otago, Dunedin 9054, New Zealand; Glycosyn (B.S.D.), Callaghan Innovation (B.S.D.), and The Ferrier Research Institute (G.F.P.), University of Wellington, Lower Hutt 5010, Victoria, New Zealand; Institut National de la Recherche Agronomique (M.B.), Nouzilly 37380, France; and School of Biological Sciences (C.S.W., D.L.H.), University of Auckland, Auckland 1142, New Zealand
| | - Massimiliano Beltramo
- Centre for Neuroendocrinology and Department of Anatomy (J.S.K., P.W.B., G.M.A.) and National School of Pharmacy (J.D.A.T.), University of Otago, Dunedin 9054, New Zealand; Glycosyn (B.S.D.), Callaghan Innovation (B.S.D.), and The Ferrier Research Institute (G.F.P.), University of Wellington, Lower Hutt 5010, Victoria, New Zealand; Institut National de la Recherche Agronomique (M.B.), Nouzilly 37380, France; and School of Biological Sciences (C.S.W., D.L.H.), University of Auckland, Auckland 1142, New Zealand
| | - Christopher S Walker
- Centre for Neuroendocrinology and Department of Anatomy (J.S.K., P.W.B., G.M.A.) and National School of Pharmacy (J.D.A.T.), University of Otago, Dunedin 9054, New Zealand; Glycosyn (B.S.D.), Callaghan Innovation (B.S.D.), and The Ferrier Research Institute (G.F.P.), University of Wellington, Lower Hutt 5010, Victoria, New Zealand; Institut National de la Recherche Agronomique (M.B.), Nouzilly 37380, France; and School of Biological Sciences (C.S.W., D.L.H.), University of Auckland, Auckland 1142, New Zealand
| | - Debbie L Hay
- Centre for Neuroendocrinology and Department of Anatomy (J.S.K., P.W.B., G.M.A.) and National School of Pharmacy (J.D.A.T.), University of Otago, Dunedin 9054, New Zealand; Glycosyn (B.S.D.), Callaghan Innovation (B.S.D.), and The Ferrier Research Institute (G.F.P.), University of Wellington, Lower Hutt 5010, Victoria, New Zealand; Institut National de la Recherche Agronomique (M.B.), Nouzilly 37380, France; and School of Biological Sciences (C.S.W., D.L.H.), University of Auckland, Auckland 1142, New Zealand
| | - Gavin F Painter
- Centre for Neuroendocrinology and Department of Anatomy (J.S.K., P.W.B., G.M.A.) and National School of Pharmacy (J.D.A.T.), University of Otago, Dunedin 9054, New Zealand; Glycosyn (B.S.D.), Callaghan Innovation (B.S.D.), and The Ferrier Research Institute (G.F.P.), University of Wellington, Lower Hutt 5010, Victoria, New Zealand; Institut National de la Recherche Agronomique (M.B.), Nouzilly 37380, France; and School of Biological Sciences (C.S.W., D.L.H.), University of Auckland, Auckland 1142, New Zealand
| | - Joel D A Tyndall
- Centre for Neuroendocrinology and Department of Anatomy (J.S.K., P.W.B., G.M.A.) and National School of Pharmacy (J.D.A.T.), University of Otago, Dunedin 9054, New Zealand; Glycosyn (B.S.D.), Callaghan Innovation (B.S.D.), and The Ferrier Research Institute (G.F.P.), University of Wellington, Lower Hutt 5010, Victoria, New Zealand; Institut National de la Recherche Agronomique (M.B.), Nouzilly 37380, France; and School of Biological Sciences (C.S.W., D.L.H.), University of Auckland, Auckland 1142, New Zealand
| | - Greg M Anderson
- Centre for Neuroendocrinology and Department of Anatomy (J.S.K., P.W.B., G.M.A.) and National School of Pharmacy (J.D.A.T.), University of Otago, Dunedin 9054, New Zealand; Glycosyn (B.S.D.), Callaghan Innovation (B.S.D.), and The Ferrier Research Institute (G.F.P.), University of Wellington, Lower Hutt 5010, Victoria, New Zealand; Institut National de la Recherche Agronomique (M.B.), Nouzilly 37380, France; and School of Biological Sciences (C.S.W., D.L.H.), University of Auckland, Auckland 1142, New Zealand
| |
Collapse
|
24
|
Xiang W, Zhang B, Lv F, Ma Y, Chen H, Chen L, Yang F, Wang P, Chu M. The Inhibitory Effects of RFamide-Related Peptide 3 on Luteinizing Hormone Release Involves an Estradiol-Dependent Manner in Prepubertal but Not in Adult Female Mice. Biol Reprod 2015; 93:30. [PMID: 26063871 DOI: 10.1095/biolreprod.115.128777] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 05/11/2015] [Indexed: 12/22/2022] Open
Abstract
The mammalian gonadotropin-inhibitory hormone (GnIH) ortholog, RFamide-related peptide (RFRP), is considered to act on gonadotropin-releasing hormone (GnRH) neurons and the pituitary to inhibit gonadotropin synthesis and release. However, there is little evidence documenting whether RFamide-related peptide 3 (RFRP-3) plays a primary role in inhibition of the hypothalamo-pituitary-gonadal (HPG) axis prior to the onset of puberty. The present study aimed to understand the functional significance of the neuropeptide on pubertal development. The developmental changes in reproductive-related gene expression at the mRNA level were investigated in the hypothalamus of female mice. The results indicated that RFRP-3 may be an endogenous inhibitory factor for the activation of the HPG axis prior to the onset of puberty. In addition, centrally administered RFRP-3 significantly suppressed plasma luteinizing hormone (LH) levels in prepubertal female mice. Surprisingly, centrally administered RFRP-3 had no effects on plasma LH levels in ovariectomized (OVX) prepubescent female mice. In contrast, RFRP-3 also inhibited plasma LH levels in OVX prepubescent female mice that were treated with 17beta-estradiol replacement. Our study also examined the effects of RFRP-3 on plasma LH release in adult female mice that were ovariectomized at dioestrus, with or without estradiol (E2). Our results showed that the inhibitory effects of RFRP-3 were independent of E2 status. Quantitative real-time PCR and immunohistochemistry analyses showed that RFRP-3 inhibited GnRH expression at both the mRNA and protein levels in the hypothalamus. These data demonstrated that RFRP-3 could effectively suppress pituitary LH release, via the inhibition of GnRH transcription and translation in prepubescent female mice, which is associated with estrogen signaling pathway and developmental stages.
Collapse
Affiliation(s)
- Wei Xiang
- College of Bioengineering, Chongqing University, Chongqing, China
| | - Baoyun Zhang
- College of Bioengineering, Chongqing University, Chongqing, China
| | - Fenglin Lv
- College of Bioengineering, Chongqing University, Chongqing, China
| | - Yunxia Ma
- College of Bioengineering, Chongqing University, Chongqing, China
| | - Hang Chen
- College of Bioengineering, Chongqing University, Chongqing, China
| | - Long Chen
- College of Bioengineering, Chongqing University, Chongqing, China
| | - Fang Yang
- College of Bioengineering, Chongqing University, Chongqing, China
| | - Pingqing Wang
- College of Bioengineering, Chongqing University, Chongqing, China
| | - Mingxing Chu
- Key Laboratory of Farm Animal Genetic Resources and Germplasm Innovation of Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
25
|
Liu X, Herbison A. Kisspeptin regulation of arcuate neuron excitability in kisspeptin receptor knockout mice. Endocrinology 2015; 156:1815-27. [PMID: 25756309 DOI: 10.1210/en.2014-1845] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The G protein-coupled receptor 54 (GPR54) is critical for kisspeptin to activate GnRH neurons to modulate fertility. However, the often mismatching distribution of kisspeptin and GPR54 in the brain suggests that kisspeptin may also act on other receptors. The arcuate nucleus (ARN) is one brain region with a very high density of kisspeptin fibers but only limited evidence for the expression of GPR54. Using acute brain slice electrophysiology in combination with Gpr54 knockout (GPR54KO) mouse models, we examined whether actions of kisspeptin in the ARN were dependent upon GPR54. Cell-attached recordings from unidentified ARN neurons in wild-type mice revealed that approximately one third of neurons were either excited or inhibited by kisspeptin in a dose-dependent manner. The responses of ARN neurons to kisspeptin were exactly the same in GPR54KO mice despite effects of kisspeptin on GnRH neurons being abolished. To evaluate whether kisspeptin may be acting through neuropeptide FF receptors, the effects of an agonist RFamide-related peptide 3 (RFRP-3) and antagonists RF9 and BIBP-3226 were evaluated. Both the excitatory and inhibitory effects of kisspeptin were mimicked by the agonist RFRP-3. RF9 itself activated ARN neurons and suppressed only the inhibitory actions of kisspeptin. BIBP-3226 suppressed kisspeptin actions in 50% of neurons. Whole-cell recordings in GPR54KO mice demonstrated that both kisspeptin and RFRP-3 acted directly on the same ARN neurons and activated the same ion channels. Together, these studies demonstrate that kisspeptin can act partly through neuropeptide FF receptors to modulate neuronal activity independent of GPR54 in the mouse brain.
Collapse
Affiliation(s)
- Xinhuai Liu
- Centre for Neuroendocrinology and Department of Physiology, University of Otago, School of Medical Sciences, Dunedin 9054, New Zealand
| | | |
Collapse
|
26
|
Sahin Z, Canpolat S, Ozcan M, Ozgocer T, Kelestimur H. Kisspeptin antagonist prevents RF9-induced reproductive changes in female rats. Reproduction 2015; 149:465-73. [PMID: 25713426 DOI: 10.1530/rep-14-0683] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The aim of this study was to determine the modulatory effects of peptide 234 (p234) (an antagonist of GPR54 receptors) on kisspeptin and RF9 (an RFamide-related peptide antagonist)-induced changes in reproductive functions and energy balance in female rats. Female Sprague-Dawley rats were weaned on postnatal day (pnd) 21. The animals were intracerebroventricularly cannulated under general anesthesia on pnd 23. Groups of female rats were injected with kisspeptin, RF9, p234, kisspeptin plus p234, or RF9 plus p234, daily. The experiments were ended on the day of first diestrus following pnd 60. Kisspeptin or RF9 alone advanced vaginal opening (VO), which was delayed by administration of kisspeptin antagonist alone. In the rats given kisspeptin plus p234 or RF9 plus p234, VO was not different from control rats. Kisspeptin and RF9 elicited significant elevations in circulating LH levels. Coadministrations of kisspeptin or RF9 with p234 decreased LH levels significantly. The use of p234 alone did not cause any significant change in LH secretion. Kisspeptin decreased both food intake and body weight while RF9 decreased only food intake without affecting body weight. The effects of kisspeptin on energy balance were also reversed by central administration of p234. In conclusion, kisspeptin antagonist, p234, modulates the effects of kisspeptin on reproductive functions and energy balance, whereas RF9 seems to exert only its effects on reproductive functions by means of GPR54 signaling in female rats.
Collapse
Affiliation(s)
- Zafer Sahin
- Department of PhysiologyFaculty of MedicineDepartment of BiophysicsMedical School, Firat University, Elazig 23119, TurkeyDepartment of PhysiologyMedical School, Inonu University, Malatya, Turkey
| | - Sinan Canpolat
- Department of PhysiologyFaculty of MedicineDepartment of BiophysicsMedical School, Firat University, Elazig 23119, TurkeyDepartment of PhysiologyMedical School, Inonu University, Malatya, Turkey
| | - Mete Ozcan
- Department of PhysiologyFaculty of MedicineDepartment of BiophysicsMedical School, Firat University, Elazig 23119, TurkeyDepartment of PhysiologyMedical School, Inonu University, Malatya, Turkey
| | - Tuba Ozgocer
- Department of PhysiologyFaculty of MedicineDepartment of BiophysicsMedical School, Firat University, Elazig 23119, TurkeyDepartment of PhysiologyMedical School, Inonu University, Malatya, Turkey
| | - Haluk Kelestimur
- Department of PhysiologyFaculty of MedicineDepartment of BiophysicsMedical School, Firat University, Elazig 23119, TurkeyDepartment of PhysiologyMedical School, Inonu University, Malatya, Turkey
| |
Collapse
|
27
|
Leon S, Tena-Sempere M. Dissecting the Roles of Gonadotropin-Inhibitory Hormone in Mammals: Studies Using Pharmacological Tools and Genetically Modified Mouse Models. Front Endocrinol (Lausanne) 2015; 6:189. [PMID: 26779117 PMCID: PMC4700143 DOI: 10.3389/fendo.2015.00189] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 12/10/2015] [Indexed: 12/24/2022] Open
Abstract
Reproduction is essential for perpetuation of the species and, hence, is controlled by a sophisticated network of regulatory factors of central and peripheral origin that integrate at the hypothalamic-pituitary-gonadal (HPG) axis. Among the central regulators of reproduction, kisspeptins, as major stimulatory drivers of gonadotropin-releasing hormone (GnRH) neurosecretion, have drawn considerable interest in the last decade. However, the dynamic, if not cyclic (in the female), nature of reproductive function and the potency of kisspeptins and other stimulatory signals of the HPG axis make tenable the existence of counterbalance inhibitory mechanisms, which may keep stimulation at check and would allow adaptation of reproductive maturation and function to different endogenous and environmental conditions. In this context, discovery of the gonadotropin-inhibitory hormone (GnIH) in birds, and its mammalian homolog, RFRP, opened up the exciting possibility that this inhibitory signal might operate centrally to suppress, directly or indirectly, GnRH/gonadotropin secretion, thus reciprocally cooperating with other stimulatory inputs in the dynamic regulation of the reproductive hypothalamic-pituitary unit. After more than 15 years of active research, the role of GnIH/RFRP in the control of the HPG axis has been documented in different species. Yet, important aspects of the physiology of this system, especially regarding its relative importance and actual roles in the control of key facets of reproductive function, remain controversial. In the present work, we aim to provide a critical review of recent developments in this area, with special attention to studies in rodent models, using pharmacological tools and functional genomics. In doing so, we intend to endow the reader with an updated view of what is known (and what is not known) about the physiological role of GnIH/RFRP signaling in the control of mammalian reproduction.
Collapse
Affiliation(s)
- Silvia Leon
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain
- Instituto de Salud Carlos III, CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain
- Instituto Maimónides de Investigación Biomédica de Córdoba/Hospital Universitario Reina Sofia (IMIBIC/HURS), Córdoba, Spain
| | - Manuel Tena-Sempere
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Córdoba, Spain
- Instituto de Salud Carlos III, CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain
- Instituto Maimónides de Investigación Biomédica de Córdoba/Hospital Universitario Reina Sofia (IMIBIC/HURS), Córdoba, Spain
- FiDiPro Program, Department of Physiology, University of Turku, Turku, Finland
- *Correspondence: Manuel Tena-Sempere,
| |
Collapse
|