51
|
Atabey M, Aykota MR, Özel Mİ, Arslan G. Short-term changes and correlations of plasma spexin, kisspeptin, and galanin levels after laparoscopic sleeve gastrectomy. Surg Today 2021; 51:651-658. [PMID: 33555434 DOI: 10.1007/s00595-021-02240-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 01/08/2021] [Indexed: 12/24/2022]
Abstract
PURPOSE To determine the circulating levels of spexin, kisspeptin, galanin, and the correlations between these peptides after laparoscopic sleeve gastrectomy (LSG). METHODS The plasma levels of the spexin, kisspeptin, and galanin and metabolic parameters (body mass index, weight loss, % excess weight loss, body fat, fasting glucose, HbA1C, and cholesterol levels) were measured (baseline, 1 month, and 3 months) and correlated in thirty adult individuals with obesity (22 female and 8 male) after LSG. RESULTS The body mass index (BMI), body fat, fasting glucose, total and low-density lipoprotein cholesterol decreased, while high-density lipoprotein cholesterol and % EWL (excess weight loss) increased at 3 months after surgery. The plasma spexin levels increased at 3 months, kisspeptin levels increased at 1 month and stabilized afterward, and galanin levels decreased at 3 months after LSG. Significant correlations were found between metabolic parameters with spexin, kisspeptin, and galanin. In addition, spexin and kisspeptin were negatively correlated with galanin, while spexin was positively correlated with kisspeptin. CONCLUSIONS The biochemical data reveal evidence that LSG causes an increase in the levels of spexin, and kisspeptin and a decrease in galanin levels. Our findings, therefore, suggest a possible interaction between these novel peptides, which have potential roles in obesity and glucose metabolism.
Collapse
Affiliation(s)
- Mustafa Atabey
- Department of General Surgery, Faculty of Medicine, University of Biruni, Istanbul, Turkey.
| | - Muhammed Raşid Aykota
- Department of General Surgery, Faculty of Medicine, University of Pamukkale, Denizli, Turkey
| | - Mehmet İlker Özel
- Department of General Surgery, Şarkışla Public Hospital, Sivas, Turkey
| | - Gökhan Arslan
- Department of Physiology, Faculty of Medicine, University of Ondokuz Mayis, Samsun, Turkey
| |
Collapse
|
52
|
Mills EG, Izzi-Engbeaya C, Abbara A, Comninos AN, Dhillo WS. Functions of galanin, spexin and kisspeptin in metabolism, mood and behaviour. Nat Rev Endocrinol 2021; 17:97-113. [PMID: 33273729 DOI: 10.1038/s41574-020-00438-1] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/21/2020] [Indexed: 02/07/2023]
Abstract
The bioactive peptides galanin, spexin and kisspeptin have a common ancestral origin and their pathophysiological roles are increasingly the subject of investigation. Evidence suggests that these bioactive peptides play a role in the regulation of metabolism, pancreatic β-cell function, energy homeostasis, mood and behaviour in several species, including zebrafish, rodents and humans. Galanin signalling suppresses insulin secretion in animal models (but not in humans), is potently obesogenic and plays putative roles governing certain evolutionary behaviours and mood modulation. Spexin decreases insulin secretion and has potent anorectic, analgesic, anxiolytic and antidepressive-like effects in animal models. Kisspeptin modulates glucose-stimulated insulin secretion, food intake and/or energy expenditure in animal models and humans. Furthermore, kisspeptin is implicated in the control of reproductive behaviour in animals, modulation of human sexual and emotional brain processing, and has antidepressive and fear-suppressing effects. In addition, galanin-like peptide is a further member of the galaninergic family that plays emerging key roles in metabolism and behaviour. Therapeutic interventions targeting galanin, spexin and/or kisspeptin signalling pathways could therefore contribute to the treatment of conditions ranging from obesity to mood disorders. However, many gaps and controversies exist, which must be addressed before the therapeutic potential of these bioactive peptides can be established.
Collapse
Affiliation(s)
- Edouard G Mills
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Chioma Izzi-Engbeaya
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, UK
| | - Ali Abbara
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, UK
| | - Alexander N Comninos
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, UK
| | - Waljit S Dhillo
- Section of Endocrinology and Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK.
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, UK.
| |
Collapse
|
53
|
Gorbunova OL, Shirshev SV. Role of Kisspeptin in Regulation of Reproductive and Immune Reactions. BIOCHEMISTRY (MOSCOW) 2021; 85:839-853. [PMID: 33045946 DOI: 10.1134/s0006297920080015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The work is focused on physiological role of the hormone kisspeptin produced by neurons of the hypothalamus anterior zone, which is a key regulator of reproduction processes. Role of the hormone in transmission of information on metabolic activity and induction of the secretion of gonadotropin-releasing hormone (GnRH) by the hypothalamus that determines gestation processes involving fertilization, placentation, fetal development, and child birth is considered. The literature data on molecular mechanisms and effects of kisspeptin on reproductive system including puberty initiation are summarized and analyzed. In addition, attention is paid to hormone-mediated changes in the cardiovascular system in pregnant women. For the first time, the review examines the effect of kisspeptin on functional activity of immune system cells presenting molecular mechanisms of the hormone signal transduction on the level of lymphoid cells that lead to the immune tolerance induction. In conclusion, a conceptual model is presented that determines the role of kisspeptin as an integrator of reproductive and immune functions during pregnancy.
Collapse
Affiliation(s)
- O L Gorbunova
- Perm Federal Research Center, Institute of Ecology and Genetics of Microorganisms, Ural Branch of the Russian Academy of Sciences, Perm, 614081, Russia.
| | - S V Shirshev
- Perm Federal Research Center, Institute of Ecology and Genetics of Microorganisms, Ural Branch of the Russian Academy of Sciences, Perm, 614081, Russia
| |
Collapse
|
54
|
Ibos KE, Bodnár É, Bagosi Z, Bozsó Z, Tóth G, Szabó G, Csabafi K. Kisspeptin-8 Induces Anxiety-Like Behavior and Hypolocomotion by Activating the HPA Axis and Increasing GABA Release in the Nucleus Accumbens in Rats. Biomedicines 2021; 9:112. [PMID: 33503835 PMCID: PMC7911394 DOI: 10.3390/biomedicines9020112] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/18/2021] [Accepted: 01/22/2021] [Indexed: 12/11/2022] Open
Abstract
Kisspeptins (Kp) are RF-amide neuropeptide regulators of the reproductive axis that also influence anxiety, locomotion, and metabolism. We aimed to investigate the effects of intracerebroventricular Kp-8 (an N-terminally truncated octapeptide) treatment in Wistar rats. Elevated plus maze (EPM), computerized open field (OF), and marble burying (MB) tests were performed for the assessment of behavior. Serum LH and corticosterone levels were determined to assess kisspeptin1 receptor (Kiss1r) activation and hypothalamic-pituitary-adrenal axis (HPA) stimulation, respectively. GABA release from the nucleus accumbens (NAc) and dopamine release from the ventral tegmental area (VTA) and NAc were measured via ex vivo superfusion. Kp-8 decreased open arm time and entries in EPM, and also raised corticosterone concentration, pointing to an anxiogenic effect. Moreover, the decrease in arm entries in EPM, the delayed increase in immobility accompanied by reduced ambulatory activity in OF, and the reduction in interactions with marbles show that Kp-8 suppressed exploratory and spontaneous locomotion. The increase in GABA release from the NAc might be in the background of hypolocomotion by inhibiting the VTA-NAc dopaminergic circuitry. As Kp-8 raised LH concentration, it could activate Kiss1r and stimulate the reproductive axis. As Kiss1r is associated with hyperlocomotion, it is more likely that neuropeptide FF receptor activation is involved in the suppression of locomotor activity.
Collapse
Affiliation(s)
- Katalin Eszter Ibos
- Department of Pathophysiology, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary; (É.B.); (Z.B.); (K.C.)
| | - Éva Bodnár
- Department of Pathophysiology, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary; (É.B.); (Z.B.); (K.C.)
| | - Zsolt Bagosi
- Department of Pathophysiology, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary; (É.B.); (Z.B.); (K.C.)
| | - Zsolt Bozsó
- Department of Medical Chemistry, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary; (Z.B.); (G.T.)
| | - Gábor Tóth
- Department of Medical Chemistry, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary; (Z.B.); (G.T.)
| | - Gyula Szabó
- Office of International Affairs, Budapest Campus, McDaniel College, H-1071 Budapest, Hungary;
| | - Krisztina Csabafi
- Department of Pathophysiology, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary; (É.B.); (Z.B.); (K.C.)
| |
Collapse
|
55
|
Musa E, Matjila M, Levitt NS. Kisspeptins and Glucose Homeostasis in Pregnancy: Implications for Gestational Diabetes Mellitus-a Review Article. Reprod Sci 2021; 29:321-327. [PMID: 33398849 DOI: 10.1007/s43032-020-00437-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 12/13/2020] [Indexed: 11/30/2022]
Abstract
Gestational diabetes mellitus (GDM) is becoming an increasingly common complication of pregnancy with the global rise of obesity. The precise pathophysiological mechanisms underpinning GDM are yet to be fully elucidated. Kisspeptin, a peptide encoded by the KISS1 gene, is mainly expressed by placental syncytiotrophoblasts during pregnancy. It is an essential ligand for kisspeptin 1 receptor (KISS1R), which is expressed by both the villous and invasive extravillous cytotrophoblast cells. Circulatory kisspeptins rise dramatically in the second and third trimester of pregnancy coinciding with the period of peak insulin resistance. Kisspeptins stimulate glucose-dependent insulin secretion and decreased plasma levels inversely correlate with markers of insulin resistance. Additionally, kisspeptins play a critical role in the regulation of appetite, energy utilisation and glucose homeostasis. GDM pregnancies have been associated with low circulatory kisspeptins, despite higher placental kisspeptin and KISS1R expression. This review evaluates the role of kisspeptin in insulin secretion, resistance and regulation of appetite as well as its implications in GDM.
Collapse
Affiliation(s)
- Ezekiel Musa
- Chronic Disease Initiative for Africa and Division of Endocrinology, Department of Medicine, University of Cape Town, Cape Town, South Africa.
| | - Mushi Matjila
- Department of Obstetrics and Gynaecology, University of Cape Town, Cape Town, South Africa
| | - Naomi S Levitt
- Chronic Disease Initiative for Africa and Division of Endocrinology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
56
|
Bédécarrats GY, Hanlon C, Tsutsui K. Gonadotropin Inhibitory Hormone and Its Receptor: Potential Key to the Integration and Coordination of Metabolic Status and Reproduction. Front Endocrinol (Lausanne) 2021; 12:781543. [PMID: 35095760 PMCID: PMC8792613 DOI: 10.3389/fendo.2021.781543] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 12/02/2021] [Indexed: 12/18/2022] Open
Abstract
Since its discovery as a novel gonadotropin inhibitory peptide in 2000, the central and peripheral roles played by gonadotropin-inhibiting hormone (GnIH) have been significantly expanded. This is highlighted by the wide distribution of its receptor (GnIH-R) within the brain and throughout multiple peripheral organs and tissues. Furthermore, as GnIH is part of the wider RF-amide peptides family, many orthologues have been characterized across vertebrate species, and due to the promiscuity between ligands and receptors within this family, confusion over the nomenclature and function has arisen. In this review, we intend to first clarify the nomenclature, prevalence, and distribution of the GnIH-Rs, and by reviewing specific localization and ligand availability, we propose an integrative role for GnIH in the coordination of reproductive and metabolic processes. Specifically, we propose that GnIH participates in the central regulation of feed intake while modulating the impact of thyroid hormones and the stress axis to allow active reproduction to proceed depending on the availability of resources. Furthermore, beyond the central nervous system, we also propose a peripheral role for GnIH in the control of glucose and lipid metabolism at the level of the liver, pancreas, and adipose tissue. Taken together, evidence from the literature strongly suggests that, in fact, the inhibitory effect of GnIH on the reproductive axis is based on the integration of environmental cues and internal metabolic status.
Collapse
Affiliation(s)
- Grégoy Y. Bédécarrats
- Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada
- *Correspondence: Grégoy Y. Bédécarrats,
| | - Charlene Hanlon
- Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada
| | - Kazuyoshi Tsutsui
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashihiroshima, Japan
| |
Collapse
|
57
|
Uenoyama Y, Nagae M, Tsuchida H, Inoue N, Tsukamura H. Role of KNDy Neurons Expressing Kisspeptin, Neurokinin B, and Dynorphin A as a GnRH Pulse Generator Controlling Mammalian Reproduction. Front Endocrinol (Lausanne) 2021; 12:724632. [PMID: 34566891 PMCID: PMC8458932 DOI: 10.3389/fendo.2021.724632] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 08/23/2021] [Indexed: 01/16/2023] Open
Abstract
Increasing evidence accumulated during the past two decades has demonstrated that the then-novel kisspeptin, which was discovered in 2001, the known neuropeptides neurokinin B and dynorphin A, which were discovered in 1983 and 1979, respectively, and their G-protein-coupled receptors, serve as key molecules that control reproduction in mammals. The present review provides a brief historical background and a summary of our recent understanding of the roles of hypothalamic neurons expressing kisspeptin, neurokinin B, and dynorphin A, referred to as KNDy neurons, in the central mechanism underlying gonadotropin-releasing hormone (GnRH) pulse generation and subsequent tonic gonadotropin release that controls mammalian reproduction.
Collapse
|
58
|
Dudás B, Merchenthaler I. Morphology and distribution of hypothalamic peptidergic systems. HANDBOOK OF CLINICAL NEUROLOGY 2021; 179:67-85. [PMID: 34225984 DOI: 10.1016/b978-0-12-819975-6.00002-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Neuropeptides participate in the regulation of numerous hypothalamic functions that are aimed for sustaining the homeostasis of the organism. These neuropeptides can act in two different levels. They can influence the release of hormones from the adenohypophysis via the portal circulation; in addition, they can act as neurotransmitters/neuromodulators modulating the functioning of numerous hypothalamic neurotransmitter systems. Indeed, most of these peptidergic systems form a complex network in the infundibular and periventricular nuclei of the human hypothalamus, communicating with each other by synaptic connections that may control fundamental physiologic functions. In the present chapter, we provide an overview of the distribution of neuropeptides in the human hypothalamus using immunohistochemistry and high-resolution, three-dimensional mapping.
Collapse
Affiliation(s)
- Bertalan Dudás
- Neuroendocrine Organization Laboratory, Lake Erie College of Osteopathic Medicine, Erie, PA, United States; Department of Anatomy, Histology and Embryology, University of Szeged, Szeged, Hungary.
| | - István Merchenthaler
- Department of Epidemiology and Public Health and of Anatomy and Neurobiology, University of Maryland Baltimore, Baltimore, MD, United States
| |
Collapse
|
59
|
D’Occhio MJ, Campanile G, Baruselli PS. Peripheral action of kisspeptin at reproductive tissues-role in ovarian function and embryo implantation and relevance to assisted reproductive technology in livestock: a review. Biol Reprod 2020; 103:1157-1170. [PMID: 32776148 PMCID: PMC7711897 DOI: 10.1093/biolre/ioaa135] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 07/23/2020] [Accepted: 08/07/2020] [Indexed: 12/13/2022] Open
Abstract
Kisspeptin (KISS1) is encoded by the KISS1 gene and was initially found to be a repressor of metastasis. Natural mutations in the KISS1 receptor gene (KISS1R) were subsequently shown to be associated with idiopathic hypothalamic hypogonadism and impaired puberty. This led to interest in the role of KISS1 in reproduction. It was established that KISS1 had a fundamental role in the control of gonadotropin releasing hormone (GnRH) secretion. KISS1 neurons have receptors for leptin and estrogen receptor α (ERα), which places KISS1 at the gateway of metabolic (leptin) and gonadal (ERα) regulation of GnRH secretion. More recently, KISS1 has been shown to act at peripheral reproductive tissues. KISS1 and KISS1R genes are expressed in follicles (granulosa, theca, oocyte), trophoblast, and uterus. KISS1 and KISS1R proteins are found in the same tissues. KISS1 appears to have autocrine and paracrine actions in follicle and oocyte maturation, trophoblast development, and implantation and placentation. In some studies, KISS1 was beneficial to in vitro oocyte maturation and blastocyst development. The next phase of KISS1 research will explore potential benefits on embryo survival and pregnancy. This will likely involve longer-term KISS1 treatments during proestrus, early embryo development, trophoblast attachment, and implantation and pregnancy. A deeper understanding of the direct action of KISS1 at reproductive tissues could help to achieve the next step change in embryo survival and improvement in the efficiency of assisted reproductive technology.
Collapse
Affiliation(s)
- Michael J D’Occhio
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| | - Giuseppe Campanile
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Pietro S Baruselli
- Department of Animal Reproduction, Faculty of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
60
|
Kisspeptin-1 regulates forebrain dopaminergic neurons in the zebrafish. Sci Rep 2020; 10:19361. [PMID: 33168887 PMCID: PMC7652893 DOI: 10.1038/s41598-020-75777-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 09/28/2020] [Indexed: 01/14/2023] Open
Abstract
The habenula is a phylogenetically conserved epithalamic structure, which conveys negative information via inhibition of mesolimbic dopamine neurons. We have previously shown the expression of kisspeptin (Kiss1) in the habenula and its role in the modulation of fear responses in the zebrafish. In this study, to investigate whether habenular Kiss1 regulates fear responses via dopamine neurons in the zebrafish, Kiss1 peptides were intracranially administered close to the habenula, and the expression of dopamine-related genes (th1, th2 and dat) were examined in the brain using real-time PCR and dopamine levels using LC–MS/MS. th1 mRNA levels and dopamine levels were significantly increased in the telencephalon 24-h and 30-min after Kiss1 administration, respectively. In fish administered with Kiss1, expression of neural activity marker gene, npas4a and kiss1 gene were significantly decreased in the ventral habenula. Application of neural tracer into the median raphe, site of habenular Kiss1 neural terminal projections showed tracer-labelled projections in the medial forebrain bundle towards the telencephalon where dopamine neurons reside. These results suggest that Kiss1 negatively regulates its own neuronal activity in the ventral habenula via autocrine action. This, in turn affects neurons of the median raphe via interneurons, which project to the telencephalic dopaminergic neurons.
Collapse
|
61
|
Beltramo M, Robert V, Decourt C. The kisspeptin system in domestic animals: what we know and what we still need to understand of its role in reproduction. Domest Anim Endocrinol 2020; 73:106466. [PMID: 32247617 DOI: 10.1016/j.domaniend.2020.106466] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 02/10/2020] [Accepted: 02/26/2020] [Indexed: 02/05/2023]
Abstract
The discovery of the kisspeptin (Kp) system stirred a burst of research in the field of reproductive neuroendocrinology. In the last 15 yr, the organization and activity of the system, including its neuroanatomical structure, its major physiological functions, and its main pharmacological properties, were outlined. To this endeavor, the use of genetic tools to delete and to restore Kp system functionality in a specific tissue was essential. At present, there is no question as to the key role of the Kp system in mammalian reproduction. However, easily applicable genetic manipulations are unavailable for domestic animals. Hence, many essential details on the physiological mechanisms underlying its action on domestic animals require further investigation. The potentially different effects of the various Kp isoforms, the precise anatomical localization of the Kp receptor, and the respective role played by the 2 main populations of Kp cells in different species are only few of the questions that remain unanswered and that will be illustrated in this review. Furthermore, the application of synthetic pharmacologic tools to manipulate the Kp system is still in its infancy but has produced some interesting results, suggesting the possibility of developing new methods to manage reproduction in domestic animals. In spite of a decade and a half of intense research effort, much work is still required to achieve a comprehensive understanding of the influence of the Kp system on reproduction. Furthermore, Kp system ramifications in other physiological functions are emerging and open new research perspectives.
Collapse
Affiliation(s)
- M Beltramo
- INRAE (CNRS, UMR7247, Université de Tours, IFCE), UMR85 Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, France.
| | - V Robert
- INRAE (CNRS, UMR7247, Université de Tours, IFCE), UMR85 Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, France
| | - C Decourt
- INRAE (CNRS, UMR7247, Université de Tours, IFCE), UMR85 Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, France
| |
Collapse
|
62
|
Babwah AV. The wonderful and masterful G protein-coupled receptor (GPCR): A focus on signaling mechanisms and the neuroendocrine control of fertility. Mol Cell Endocrinol 2020; 515:110886. [PMID: 32574585 DOI: 10.1016/j.mce.2020.110886] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/08/2020] [Accepted: 05/20/2020] [Indexed: 12/18/2022]
Abstract
Human GnRH deficiency, both clinically and genetically, is a heterogeneous disorder comprising of congenital GnRH deficiency with anosmia (Kallmann syndrome), or with normal olfaction [normosmic idiopathic hypogonadotropic hypogonadism (IHH)], and adult-onset hypogonadotropic hypogonadism. Our understanding of the neural mechanisms underlying GnRH secretion and GnRH signaling continues to increase at a rapid rate and strikingly, the heterotrimeric guanine nucleotide-binding protein (G protein)-coupled receptors (GPCRs) continue to emerge as essential players in these processes. GPCRs were once viewed as binary on-off switches, where in the "on" state they are bound to their Gα protein, but now we understand that view is overly simplistic and does not adequately characterize GPCRs. Instead, GPCRs have emerged as masterful signaling molecules exploiting different physical conformational states of itself to elicit an array of downstream signaling events via their G proteins and the β-arrestins. The "one receptor-multiple signaling conformations" model is likely an evolved strategy that can be used to our advantage as researchers have shown that targeting specific receptor conformations via biased ligands is proving to be a powerful tool in the effective treatment of human diseases. Can biased ligands be used to selectively modulate signaling by GPCR regulators of the neuroendocrine axis in the treatment of IHH? As discussed in this review, the grand possibility exists. However, while we are still very far from developing these treatments, this exciting likelihood can happen through a much greater mechanistic understanding of how GPCRs signal within the cell.
Collapse
Affiliation(s)
- Andy V Babwah
- Department of Pediatrics, Laboratory of Human Growth and Reproductive Development, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States; Child Health Institute of New Jersey, New Brunswick, NJ, United States.
| |
Collapse
|
63
|
Abstract
The significance of KISS1 goes beyond its original discovery as a metastasis suppressor. Its function as a neuropeptide involved in diverse physiologic processes is more well studied. Enthusiasm regarding KISS1 has cumulated in clinical trials in multiple fields related to reproduction and metabolism. But its cancer therapeutic space is unsettled. This review focuses on collating data from cancer and non-cancer fields in order to understand shared and disparate signaling that might inform clinical development in the cancer therapeutic and biomarker space. Research has focused on amino acid residues 68-121 (kisspeptin 54), binding to the KISS1 receptor and cellular responses. Evidence and counterevidence regarding this canonical pathway require closer look at the covariates so that the incredible potential of KISS1 can be realized.
Collapse
Affiliation(s)
- Thuc Ly
- Department of Cancer Biology, Kansas University Medical Center, 3901 Rainbow Blvd. - MS1071, Kansas City, KS, 66160, USA
| | - Sitaram Harihar
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603203, India
| | - Danny R Welch
- Department of Cancer Biology, Kansas University Medical Center, 3901 Rainbow Blvd. - MS1071, Kansas City, KS, 66160, USA.
- University of Kansas Cancer Center, 3901 Rainbow Blvd., Kansas City, KS, 66160, USA.
| |
Collapse
|
64
|
Seshan G, Kanagasabai S, Ananthasri S, Kannappan B, Suvitha A, Jaimohan SM, Kanagaraj S, Kothandan G. Insights of structure-based pharmacophore studies and inhibitor design against Gal3 receptor through molecular dynamics simulations. J Biomol Struct Dyn 2020; 39:6987-6999. [PMID: 32772816 DOI: 10.1080/07391102.2020.1804452] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Our present work studies the structure-based pharmacophore modeling and designing inhibitor against Gal3 receptor through molecular dynamics (MD) simulations extensively. Pharmacophore models play a key role in computer-aided drug discovery like in the case of virtual screening of chemical databases, de novo drug design and lead optimization. Structure-based methods for developing pharmacophore models are important, and there have been a number of studies combining such methods with the use of MD simulations to model protein's flexibility. The two potential antagonists SNAP 37889 and SNAP 398299 were docked and simulated for 250 ns and the results are analyzed and carried for the structure-based pharmacophore studies. This helped in identification of the subtype selectivity of the binding sites of the Gal3 receptor. Our work mainly focuses on identifying these binding site residues and to design more potent inhibitors compared to the previously available inhibitors through pharmacophore models. The study provides crucial insight into the binding site residues Ala2, Asp3, Ala4, Gln5, Phe24, Gln79, Ala80, Ile82, Tyr83, Trp88, His99, Ile102, Tyr103, Met106, Tyr157, Tyr161, Pro174, Trp176, Arg181, Ala183, Leu184, Asp185, Thr188, Trp248, His251, His252, Ile255, Leu256, Phe258, Trp259, Tyr270, Arg273, Leu274 and His277, which plays a significant role in the conformational changes of the receptor and helps to understand the inhibition mechanism. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Gunalan Seshan
- Biopolymer Modelling Laboratory, Centre of Advanced Study in Crystallography and Biophysics, University of Madras, Guindy Campus, Chennai, Tamil Nadu, India
| | - Somarathinam Kanagasabai
- Biopolymer Modelling Laboratory, Centre of Advanced Study in Crystallography and Biophysics, University of Madras, Guindy Campus, Chennai, Tamil Nadu, India
| | - Sailapathi Ananthasri
- Biopolymer Modelling Laboratory, Centre of Advanced Study in Crystallography and Biophysics, University of Madras, Guindy Campus, Chennai, Tamil Nadu, India
| | - Balaji Kannappan
- National Research Center for Dementia, Department of Life Science, Chosun University, Gwangju, South Korea
| | - A Suvitha
- Department of Physics, CMR Institute of Technology, Bangalore, Karnataka, India
| | - S M Jaimohan
- Advanced Materials Laboratory, CSIR-Central Leather Research Institute, Chennai, Tamil Nadu, India
| | - Sekar Kanagaraj
- Laboratory for Structural Biology and Biocomputing, Department of Computational and Data Sciences, Indian Institute of Science, Bangalore, Karnataka, India
| | - Gugan Kothandan
- Biopolymer Modelling Laboratory, Centre of Advanced Study in Crystallography and Biophysics, University of Madras, Guindy Campus, Chennai, Tamil Nadu, India
| |
Collapse
|
65
|
Hu K, Sun W, Li Y, Zhang B, Zhang M, Guo C, Chang H, Wang X. Study on the Mechanism of Sarsasapogenin in Treating Precocious Puberty by Regulating the HPG Axis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2020; 2020:1978043. [PMID: 32831859 PMCID: PMC7426762 DOI: 10.1155/2020/1978043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 07/13/2020] [Indexed: 12/18/2022]
Abstract
The present study aims to investigate the effects and mechanisms of sarsasapogenin resistance to precocious puberty. Female Sprague Dawley rats were divided into a normal (N) group, model (M) group, leuprolide (L) group, and sarsasapogenin (Sar) group. Rats at 5 days of age were given a single subcutaneous injection of 300 micrograms of danazol to establish the precocious puberty model. After 10 days of modeling, drug intervention was started. The development of the uterus and ovary was observed by hematoxylin and eosin (HE) staining. The levels of the serum luteinizing hormone (LH), follicle-stimulating hormone (FSH), and estradiol (E2) were determined by radioimmunoassay. Also, the expressions of the hypothalamic gonadotropin releasing hormone (GnRH), Kiss-1, G protein-coupled receptor 54 (GPR54), and pituitary gonadotropin releasing hormone receptor (GnRH-R) were detected by RT-PCR. The results showed that compared with the model group, sarsasapogenin could significantly delay the opening time of vaginal, decreased uterine and ovarian coefficients, and reduced uterine wall thickness. Moreover, it can significantly downregulate the levels of serum hormones and reduce the expression of GnRH, GnRH-R, and kiss-1. In summary, our results indicate that sarsasapogenin can regulate the HPG axis through the kiss-1/GPR54 system for therapeutic precocious puberty.
Collapse
Affiliation(s)
- Kaili Hu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100102, China
| | - Wenyan Sun
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100102, China
| | - Yu Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100102, China
| | - Bo Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100102, China
| | - Meng Zhang
- Beijing Children's Hospital, Capital Medical University, National Center for Children Health, Beijing 100045, China
| | - Chunyan Guo
- Beijing Children's Hospital, Capital Medical University, National Center for Children Health, Beijing 100045, China
| | - HongSheng Chang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100102, China
| | - Xiaoling Wang
- Beijing Children's Hospital, Capital Medical University, National Center for Children Health, Beijing 100045, China
| |
Collapse
|
66
|
Duan C, Allard J. Gonadotropin-releasing hormone neuron development in vertebrates. Gen Comp Endocrinol 2020; 292:113465. [PMID: 32184073 DOI: 10.1016/j.ygcen.2020.113465] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 02/26/2020] [Accepted: 03/12/2020] [Indexed: 11/21/2022]
Abstract
Gonadotropin-releasing hormone (GnRH) neurons are master regulators of the reproductive axis in vertebrates. During early mammalian embryogenesis, GnRH1 neurons emerge in the nasal/olfactory placode. These neurons undertake a long-distance migration, moving from the nose to the preoptic area and hypothalamus. While significant advances have been made in understanding the functional importance of the GnRH1 neurons in reproduction, where GnRH1 neurons come from and how are they specified during early development is still under debate. In addition to the GnRH1 gene, most vertebrate species including humans have one or two additional GnRH genes. Compared to the GnRH1 neurons, much less is known about the development and regulation of GnRH2 neuron and GnRH3 neurons. The objective of this article is to review what is currently known about GnRH neuron development. We will survey various cell autonomous and non-autonomous factors implicated in the regulation of GnRH neuron development. Finally, we will discuss emerging tools and new approaches to resolve open questions pertaining to GnRH neuron development.
Collapse
Affiliation(s)
- Cunming Duan
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, United States.
| | - John Allard
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, United States
| |
Collapse
|
67
|
Hu KL, Chang HM, Zhao HC, Yu Y, Li R, Qiao J. Potential roles for the kisspeptin/kisspeptin receptor system in implantation and placentation. Hum Reprod Update 2020; 25:326-343. [PMID: 30649364 PMCID: PMC6450039 DOI: 10.1093/humupd/dmy046] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 10/19/2018] [Accepted: 12/09/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Initially identified as suppressors of metastasis in various types of cancer, kisspeptins are a family of neuropeptides that are key regulators of the mammalian reproductive axis. Accumulating evidence has shown that kisspeptin is able to control both the pulsatile and surge GnRH release, playing fundamental roles in female reproduction, which include the secretion of gonadotropins, puberty onset, brain sex differentiation, ovulation and the metabolic regulation of fertility. Furthermore, recent studies have demonstrated the involvement of the kisspeptin system in the processes of implantation and placentation. This review summarizes the current knowledge of the pathophysiological role and utility of these local placental regulatory factors as potential biomarkers during the early human gestation. OBJECTIVE AND RATIONALE A successful pregnancy, from the initiation of embryo implantation to parturition, is a complex process that requires the orchestration of a series of events. This review aims to concisely summarize what is known about the role of the kisspeptin system in implantation, placentation, early human pregnancy and pregnancy-related disorders, and to develop strategies for predicting, diagnosing and treating these abnormalities. SEARCH METHODS Using the PubMed and Google Scholar databases, we performed comprehensive literature searches in the English language describing the advancement of kisspeptins and the kisspeptin receptor (KISS1R) in implantation, placentation and early pregnancy in humans, since its initial identification in 1996 and ending in July 2018. OUTCOMES Recent studies have shown the coordinated spatial and temporal expression patterns of kisspeptins and KISS1R during human pregnancy. The experimental data gathered recently suggest putative roles of kisspeptin signaling in the regulation of trophoblast invasion, embryo implantation, placentation and early pregnancy. Dysregulation of the kisspeptin system may negatively affect the processes of implantation as well as placentation. Clinical studies indicate that the circulating levels of kisspeptins or the expression levels of kisspeptin/KISS1R in the placental tissues may be used as potential diagnostic markers for women with miscarriage and gestational trophoblastic neoplasia. WIDER IMPLICATIONS Comprehensive research on the pathophysiological role of the kisspeptin/KISS1R system in implantation and placentation will provide a dynamic and powerful approach to understanding the processes of early pregnancy, with potential applications in observational and analytic screening as well as the diagnosis, prognosis and treatment of implantation failure and early pregnancy-related disorders.
Collapse
Affiliation(s)
- Kai-Lun Hu
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Department of Obstetrics and Gynecology, Ministry of Education, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
| | - Hsun-Ming Chang
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Department of Obstetrics and Gynecology, Ministry of Education, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
| | - Hong-Cui Zhao
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Department of Obstetrics and Gynecology, Ministry of Education, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
| | - Yang Yu
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Department of Obstetrics and Gynecology, Ministry of Education, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China.,National Clinical Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Rong Li
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Department of Obstetrics and Gynecology, Ministry of Education, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China.,National Clinical Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Jie Qiao
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Department of Obstetrics and Gynecology, Ministry of Education, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China.,National Clinical Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| |
Collapse
|
68
|
Harihar S, Ray S, Narayanan S, Santhoshkumar A, Ly T, Welch DR. Role of the tumor microenvironment in regulating the anti-metastatic effect of KISS1. Clin Exp Metastasis 2020; 37:209-223. [PMID: 32088827 PMCID: PMC7339126 DOI: 10.1007/s10585-020-10030-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 02/19/2020] [Indexed: 12/29/2022]
Abstract
KISS1, a metastasis suppressor gene, has been shown to block metastasis without affecting primary tumor formation. Loss of KISS1 leads to invasion and metastasis in multiple cancers, which is the leading cause of cancer morbidity and mortality. The discovery of KISS1 has provided a ray of hope for early clinical diagnosis and for designing effective treatments targeting metastatic cancer. However, this goal requires greater holistic understanding of its mechanism of action. In this review, we go back into history and highlight some key developments, from the discovery of KISS1 to its role in regulating multiple physiological processes including cancer. We discuss key emerging roles for KISS1, specifically interactions with tissue microenvironment to promote dormancy and regulation of tumor cell metabolism, acknowledged as some of the key players in tumor progression and metastasis. We finally discuss strategies whereby KISS1 might be exploited clinically to treat metastasis.
Collapse
Affiliation(s)
- Sitaram Harihar
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India.
| | - Srijit Ray
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India
| | - Samyukta Narayanan
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India
| | - Anirudh Santhoshkumar
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India
| | - Thuc Ly
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS, 66160, USA
- The University Kansas Cancer Center, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS, 66160, USA
| | - Danny R Welch
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS, 66160, USA
- The University Kansas Cancer Center, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS, 66160, USA
| |
Collapse
|
69
|
Immunoreactivity of Kisspeptin and Kisspeptin Receptor in Eutopic and Ectopic Endometrial Tissue of Women With and Without Endometriosis. Reprod Sci 2020; 27:1731-1741. [PMID: 32072605 DOI: 10.1007/s43032-020-00167-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 01/13/2020] [Indexed: 01/10/2023]
Abstract
Endometriosis is characterized by the presence of ectopic endometrial tissues. Mechanisms of tissue dissemination in endometriosis may be similar to those involved in tumor metastasis. We hypothesize that dysregulation of kisspeptin (KISS1), a metastasis suppressor in endometrial carcinoma, may contribute to the pathogenesis of endometriosis. In this study, we characterized the immunoreactivity of kisspeptin and its receptor, KISS1R, in eutopic and ectopic endometrial tissue of women with and without endometriosis, in proliferative and secretory menstrual cycle phases. Immunohistochemistry was performed using KISS1 and KISS1R antibodies on samples from women with (n = 35) and without (n = 14) endometriosis. Samples from women with endometriosis included eutopic endometrium (n = 20) samples, superficial endometriotic implants (SUP, n = 10) deep infiltrating endometriotic implants (DIE, n = 15), and ovarian endometriomas (OMA, n = 15). Immunoreactivity was quantified using histoscores. KISS1 and KISS1R immunoreactivity was significantly lower in eutopic endometrial stroma of women with versus without endometriosis, regardless of the menstrual cycle phase (P = 0.001 and P = 0.015 respectively). In endometriotic implants, KISS1 levels were significantly lower in both glandular and stromal components of DIE (P < 0.01) and OMA (P < 0.01) compared to SUP. KISS1R immunoreactivity was lower in the glandular component of OMA (P = 0.035) compared to SUP. KISS1 and KISS1R levels are lower in eutopic endometrial stroma from women with versus without endometriosis, consistent with a role for decreased KISS1 expression in the pathogenesis of endometriosis. As deeply invasive lesions showed lower KISS1 levels than superficial lesions, downregulation of KISS1 levels may contribute to implant invasiveness.
Collapse
|
70
|
Blanco AM. Hypothalamic- and pituitary-derived growth and reproductive hormones and the control of energy balance in fish. Gen Comp Endocrinol 2020; 287:113322. [PMID: 31738909 DOI: 10.1016/j.ygcen.2019.113322] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 10/20/2019] [Accepted: 11/12/2019] [Indexed: 02/07/2023]
Abstract
Most endocrine systems in the body are influenced by the hypothalamic-pituitary axis. Within this axis, the hypothalamus delivers precise signals to the pituitary gland, which in turn releases hormones that directly affect target tissues including the liver, thyroid gland, adrenal glands and gonads. This action modulates the release of additional hormones from the sites of action, regulating key physiological processes, including growth, metabolism, stress and reproduction. Pituitary hormones are released by five distinct hormone-producing cell types: somatotropes (which produce growth hormone), thyrotropes (thyrotropin), corticotropes (adrenocorticotropin), lactotropes (prolactin) and gonadotropes (follicle stimulating hormone and luteinizing hormone), each modulated by specific hypothalamic signals. This careful and distinct organization of the hypothalamo-pituitary axis has been classically associated with the existence of many lineal axes (e.g., the hypothalamic-pituitary-gonadal axis) in charge of the control of the different physiological processes. While this traditional concept is valid, it is becoming apparent that hormones produced by the hypothalamo-pituitary axis have diverse effects. For instance, gonadotropin-releasing hormone II has been associated with a suppressive effect on food intake in fish. Likewise, growth hormone has been shown to influence appetite, swimming activity and aggressive behavior in fish. This review will focus on the hypothalamic and pituitary hormones classically involved in regulating growth and reproduction, and will attempt to provide a general overview of the current knowledge on their actions on energy balance and appetite in fish. It will also give a brief perspective of the role of some of these peptides in integrating feeding, metabolism, growth and reproduction.
Collapse
Affiliation(s)
- Ayelén M Blanco
- Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía and Centro de Investigación Mariña, Universidade de Vigo, Vigo, Pontevedra, Spain; Laboratory of Integrative Neuroendocrinology, Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.
| |
Collapse
|
71
|
El-Sherry TM, Abdel-Ghani MA, Mahmoud GB, Ezzat AA. Kisspeptin injection improved the semen characteristics and sperm rheotaxis in Ossimi ram. Reprod Domest Anim 2020; 55:240-247. [PMID: 31880370 DOI: 10.1111/rda.13613] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 12/08/2019] [Indexed: 11/30/2022]
Abstract
The aim of the present study was to investigate the effect of kisspeptin-10 (Kp10) injection on semen characteristics, testosterone (T) production and sperm rheotaxis using microfluidic devices in immature ram. Computer-assisted sperm analysis (CASA) with controlled flow velocity was used to explore the kinetic parameters of sperm and positive rheotaxis (PR %). PR % was defined as the number of PR sperms over the number of motile sperms. Healthy Ossimi rams were randomly divided into two groups; a saline-treated control group and Kp10-treated one (5 µg/kg body weight). Treatments were given by intramuscular injection once a week for 1 month. After 1 month, the semen was collected and evaluated weekly for 6 weeks, while the blood samples were collected every 2 weeks for the next 8 weeks. Semen properties were significantly affected by Kp10 injection (p < .01). The Kp10 increased the volume, sperm concentration and percentages of live sperm compared with those of control. Additionally, sperm trajectories and rheotaxis get improved by the injection of Kp10 with time. Furthermore, kisspeptin improved the secretion of testosterone levels throughout the period of study. In conclusion, injections of the Kp10 had a positive impact on semen characteristics as well as improved sperm rheotaxis of Ossimi rams in subtropics.
Collapse
Affiliation(s)
- Timor M El-Sherry
- Department of Theriogenology, Faculty of Veterinary Medicine, Assuit University, Assuit, Egypt
| | - Mohammed A Abdel-Ghani
- Department of Theriogenology, Faculty of Veterinary Medicine, Assuit University, Assuit, Egypt
| | - Gamal B Mahmoud
- Department of Animal Production, Faculty of Agriculture, Assiut University, Assuit, Egypt
| | - Ahmed A Ezzat
- Department of Biology, College of Science, King Khalid Unversity, Abha, Saudi Arabia.,Department of Theriogenology, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| |
Collapse
|
72
|
Wang L, Moenter SM. Differential Roles of Hypothalamic AVPV and Arcuate Kisspeptin Neurons in Estradiol Feedback Regulation of Female Reproduction. Neuroendocrinology 2020; 110:172-184. [PMID: 31466075 PMCID: PMC7047625 DOI: 10.1159/000503006] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 08/28/2019] [Indexed: 11/19/2022]
Abstract
Mammalian reproductive function includes puberty onset and completion, reproductive cyclicity, steroidogenesis, gametogenesis, fertilization, pregnancy, and lactation; all are indispensable to perpetuate species. Reproductive cycles are critical for providing the hormonal milieu needed for follicular development and maturation of eggs, but cycles, in and of themselves, do not guarantee ovulation will occur. Here, we review the roles in female reproductive neuroendocrine function of two hypothalamic populations that produce the neuropeptide kisspeptin, demonstrating distinct roles in maintaining cycles and ovulation.
Collapse
Affiliation(s)
- Luhong Wang
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Suzanne M Moenter
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA,
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA,
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA,
| |
Collapse
|
73
|
Dufour S, Quérat B, Tostivint H, Pasqualini C, Vaudry H, Rousseau K. Origin and Evolution of the Neuroendocrine Control of Reproduction in Vertebrates, With Special Focus on Genome and Gene Duplications. Physiol Rev 2019; 100:869-943. [PMID: 31625459 DOI: 10.1152/physrev.00009.2019] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In humans, as in the other mammals, the neuroendocrine control of reproduction is ensured by the brain-pituitary gonadotropic axis. Multiple internal and environmental cues are integrated via brain neuronal networks, ultimately leading to the modulation of the activity of gonadotropin-releasing hormone (GnRH) neurons. The decapeptide GnRH is released into the hypothalamic-hypophysial portal blood system and stimulates the production of pituitary glycoprotein hormones, the two gonadotropins luteinizing hormone and follicle-stimulating hormone. A novel actor, the neuropeptide kisspeptin, acting upstream of GnRH, has attracted increasing attention in recent years. Other neuropeptides, such as gonadotropin-inhibiting hormone/RF-amide related peptide, and other members of the RF-amide peptide superfamily, as well as various nonpeptidic neuromediators such as dopamine and serotonin also provide a large panel of stimulatory or inhibitory regulators. This paper addresses the origin and evolution of the vertebrate gonadotropic axis. Brain-pituitary neuroendocrine axes are typical of vertebrates, the pituitary gland, mediator and amplifier of brain control on peripheral organs, being a vertebrate innovation. The paper reviews, from molecular and functional perspectives, the evolution across vertebrate radiation of some key actors of the vertebrate neuroendocrine control of reproduction and traces back their origin along the vertebrate lineage and in other metazoa before the emergence of vertebrates. A focus is given on how gene duplications, resulting from either local events or from whole genome duplication events, and followed by paralogous gene loss or conservation, might have shaped the evolutionary scenarios of current families of key actors of the gonadotropic axis.
Collapse
Affiliation(s)
- Sylvie Dufour
- Muséum National d'Histoire Naturelle, Biology of Aquatic Organisms and Ecosystems, CNRS, IRD, Sorbonne Université, Université Caen Normandie, Université des Antilles, Paris, France; Université Paris Diderot, Sorbonne Paris Cite, Biologie Fonctionnelle et Adaptative, Paris, France; INSERM U1133, Physiologie de l'axe Gonadotrope, Paris, France; Muséum National d'Histoire Naturelle, Physiologie Moléculaire et Adaptation, Muséum National d'Histoire Naturelle, Paris, France; Université Paris-Saclay, Université Paris-Sud, CNRS, Paris-Saclay Institute of Neuroscience (UMR 9197), Gif-sur-Yvette, France; and Université de Rouen Normandie, Rouen, France
| | - Bruno Quérat
- Muséum National d'Histoire Naturelle, Biology of Aquatic Organisms and Ecosystems, CNRS, IRD, Sorbonne Université, Université Caen Normandie, Université des Antilles, Paris, France; Université Paris Diderot, Sorbonne Paris Cite, Biologie Fonctionnelle et Adaptative, Paris, France; INSERM U1133, Physiologie de l'axe Gonadotrope, Paris, France; Muséum National d'Histoire Naturelle, Physiologie Moléculaire et Adaptation, Muséum National d'Histoire Naturelle, Paris, France; Université Paris-Saclay, Université Paris-Sud, CNRS, Paris-Saclay Institute of Neuroscience (UMR 9197), Gif-sur-Yvette, France; and Université de Rouen Normandie, Rouen, France
| | - Hervé Tostivint
- Muséum National d'Histoire Naturelle, Biology of Aquatic Organisms and Ecosystems, CNRS, IRD, Sorbonne Université, Université Caen Normandie, Université des Antilles, Paris, France; Université Paris Diderot, Sorbonne Paris Cite, Biologie Fonctionnelle et Adaptative, Paris, France; INSERM U1133, Physiologie de l'axe Gonadotrope, Paris, France; Muséum National d'Histoire Naturelle, Physiologie Moléculaire et Adaptation, Muséum National d'Histoire Naturelle, Paris, France; Université Paris-Saclay, Université Paris-Sud, CNRS, Paris-Saclay Institute of Neuroscience (UMR 9197), Gif-sur-Yvette, France; and Université de Rouen Normandie, Rouen, France
| | - Catherine Pasqualini
- Muséum National d'Histoire Naturelle, Biology of Aquatic Organisms and Ecosystems, CNRS, IRD, Sorbonne Université, Université Caen Normandie, Université des Antilles, Paris, France; Université Paris Diderot, Sorbonne Paris Cite, Biologie Fonctionnelle et Adaptative, Paris, France; INSERM U1133, Physiologie de l'axe Gonadotrope, Paris, France; Muséum National d'Histoire Naturelle, Physiologie Moléculaire et Adaptation, Muséum National d'Histoire Naturelle, Paris, France; Université Paris-Saclay, Université Paris-Sud, CNRS, Paris-Saclay Institute of Neuroscience (UMR 9197), Gif-sur-Yvette, France; and Université de Rouen Normandie, Rouen, France
| | - Hubert Vaudry
- Muséum National d'Histoire Naturelle, Biology of Aquatic Organisms and Ecosystems, CNRS, IRD, Sorbonne Université, Université Caen Normandie, Université des Antilles, Paris, France; Université Paris Diderot, Sorbonne Paris Cite, Biologie Fonctionnelle et Adaptative, Paris, France; INSERM U1133, Physiologie de l'axe Gonadotrope, Paris, France; Muséum National d'Histoire Naturelle, Physiologie Moléculaire et Adaptation, Muséum National d'Histoire Naturelle, Paris, France; Université Paris-Saclay, Université Paris-Sud, CNRS, Paris-Saclay Institute of Neuroscience (UMR 9197), Gif-sur-Yvette, France; and Université de Rouen Normandie, Rouen, France
| | - Karine Rousseau
- Muséum National d'Histoire Naturelle, Biology of Aquatic Organisms and Ecosystems, CNRS, IRD, Sorbonne Université, Université Caen Normandie, Université des Antilles, Paris, France; Université Paris Diderot, Sorbonne Paris Cite, Biologie Fonctionnelle et Adaptative, Paris, France; INSERM U1133, Physiologie de l'axe Gonadotrope, Paris, France; Muséum National d'Histoire Naturelle, Physiologie Moléculaire et Adaptation, Muséum National d'Histoire Naturelle, Paris, France; Université Paris-Saclay, Université Paris-Sud, CNRS, Paris-Saclay Institute of Neuroscience (UMR 9197), Gif-sur-Yvette, France; and Université de Rouen Normandie, Rouen, France
| |
Collapse
|
74
|
Ke R, Ma X, Lee LTO. Understanding the functions of kisspeptin and kisspeptin receptor (Kiss1R) from clinical case studies. Peptides 2019; 120:170019. [PMID: 30339828 DOI: 10.1016/j.peptides.2018.09.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 09/21/2018] [Accepted: 09/21/2018] [Indexed: 10/28/2022]
Abstract
It is widely acknowledged that kisspeptin and its receptor Kiss1R play central regulatory roles in the hypothalamus-pituitary-gonad (HPG) axis and reproduction. Mutations of KISS1 and KISS1R lead to disorders associated with pubertal development, such as central precocious puberty (CPP) and idiopathic hypogonadotropic hypogonadism (IHH). This review focuses on KISS1 and KISS1R mutations found in CPP and IHH and its purposes are twofold: Firstly, based on the mutations found in KISS1 and KISS1R, this review provides insights into the precise mechanism of kisspeptin and the kisspeptin/Kiss1R pathway in the reproductive axis and in puberty. Secondly, G protein-coupled receptors (GPCRs) are known to share highly conserved structural motifs; therefore, knowledge of mutations found at different structural domains of Kiss1R in the diseased state, and how they affect Kiss1R function can be used to decipher GPCR domain function.
Collapse
Affiliation(s)
- Ran Ke
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Xin Ma
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Leo T O Lee
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau, China.
| |
Collapse
|
75
|
Bayerl DS, Klampfl SM, Bosch OJ. More than reproduction: Central gonadotropin-releasing hormone antagonism decreases maternal aggression in lactating rats. J Neuroendocrinol 2019; 31:e12709. [PMID: 30882966 DOI: 10.1111/jne.12709] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 01/29/2019] [Accepted: 03/13/2019] [Indexed: 12/20/2022]
Abstract
Gonadotropin-releasing hormone (GnRH) is a major regulator and activator of the hypothalamic-pituitary-gonadal axis. Many studies have demonstrated the importance of GnRH in reproduction and sexual behaviour. However, to date, only a single study shows an involvement of GnRH in maternal behaviour where a 30% reduction of GnRH neurones abolishes a mother's motivation to retrieve pups. On this basis, we aimed to investigate the effects of acute central GnRH receptor blockade in lactating rats on maternal care under non-stress and stress conditions, maternal motivation in the pup retrieval test, maternal anxiety on the elevated plus maze, and maternal aggression in the maternal defence test. We found that acute central infusion of a GnRH antagonist ([d-Phe2,6 ,Pro3 ]-luteinising hormone-releasing hormone; 0.5 ng 5 μL-1 ) impaired a mother's attack behaviour against a female intruder rat during the maternal defence test compared to vehicle controls. However, in contrast to the previous study on reduced GnRH neurones, acute central GnRH antagonism did not affect pup retrieval, nor any other parameter of maternal behaviour or maternal anxiety. Taken together, GnRH receptor activation is mandatory for protection of the offspring. These findings shed new light on GnRH as a neuropeptide acting not exclusively on the reproductive axis but, additionally, on maternal behaviour including pup retrieval and maternal aggression.
Collapse
Affiliation(s)
- Doris S Bayerl
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| | - Stefanie M Klampfl
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| | - Oliver J Bosch
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| |
Collapse
|
76
|
Wu F, Zhang W, Song QQ, Li HH, Xu MS, Liu GL, Zhang JZ. Association analysis of polymorphisms of G protein-coupled receptor 54 gene exons with reproductive traits in Jiaxing Black sows. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2019; 32:1104-1111. [PMID: 30744352 PMCID: PMC6599949 DOI: 10.5713/ajas.18.0827] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/05/2018] [Accepted: 12/18/2018] [Indexed: 11/27/2022]
Abstract
OBJECTIVE The aim of this study was to detect single nucleotide polymorphisms (SNP) of G protein-coupled receptor 54 (GPR54) gene and explore association of this candidate gene with reproductive traits in Jiaxing Black sows. METHODS Six pairs of primers of the gene were designed to amplify all exons thus sequences of which were detected by means of direct sequencing and then SNP loci were scanned. The effects of SNPs on total number of piglets born (TNB), number of piglets born alive (NBA), number of still born piglets (NSB), and litter weight at birth (LWB) of Jiaxing Black sows were analyzed. RESULTS Three SNP loci, including T3739C, C3878T and T6789C, were identified via comparison of sequencing and two genotypes (AB, BB) at each SNP site were observed. T3739C resulted in the change of amino acid (Leu→Pro) in corresponding protein, and C3878T resulted in synonymous mutation (Ile→Ile). Statistical results demonstrated that allele B was the preponderant allele at the three SNP loci and Genotype BB was the preponderant genotype. Meanwhile, Chi-Square test of these three SNPs indicated that all mutation sites fitted in Hardy-Weinberg equilibrium (p>0.05). For GPR54-T3739C locus, Jiaxing Black sows with genotype BB had 1.23 TNB and 1.28 NBA (p<0.01) that were more than those with genotype AB, respectively. Jiaxing Black sows that had the first two parities with genotype BB had additional 2.23 TNB, 2.27 NBA (p<0.01), and 1.94 LWB (p<0.05) compared to those with genotype AB, respectively. However, for other two loci, no significant difference was found between TNB, NBA, NSB, and LWB, and different genotypes of Jiaxing Black sows. CONCLUSION In conclusion, the polymorphisms of GPR54-T3739C locus were significantly associated to TNB, NBA, and LWB and could be used as a potential genetic marker to improve reproductive function of Jiaxing black sows.
Collapse
Affiliation(s)
- Fen Wu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058,
China
| | - Wei Zhang
- College of Animal Sciences, Zhejiang University, Hangzhou 310058,
China
| | - Qian-Qian Song
- College of Animal Sciences, Zhejiang University, Hangzhou 310058,
China
| | - Hai-Hong Li
- Zhejiang Qinglian Food Company Limited, Jiaxing 314000,
China
| | - Ming-Shu Xu
- Zhejiang Qinglian Food Company Limited, Jiaxing 314000,
China
| | - Guo-Liang Liu
- Zhejiang Qinglian Food Company Limited, Jiaxing 314000,
China
| | - Jin-Zhi Zhang
- College of Animal Sciences, Zhejiang University, Hangzhou 310058,
China
| |
Collapse
|
77
|
Feng T, Bai JH, Xu XL, Liu Y. Kisspeptin and its Effect on Mammalian Spermatogensis. Curr Drug Metab 2019; 20:9-14. [PMID: 29380696 DOI: 10.2174/1389200219666180129112406] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 11/20/2017] [Accepted: 12/03/2017] [Indexed: 11/22/2022]
Abstract
BACKGROUND Kisspeptin and its receptor, GPR54, are regarded as key regulators of and catalysts for male puberty onset, and also fundamental gatekeepers of spermatogenesis in mammals. Consequently, the loss function of kisspeptin or GPR54 leads to a symptom of Hypogonadotropic Hypogonadism (HH) in human and HH accompanied by lower gonadotrophic hormone levels, smaller testes, impaired spermatogenesis and abnormal sexual maturation in mice. Besides its well-recognized functions in hypothalamus before and during puberty, accumulating data strongly support kisspeptin production in testis, and participation in somatic and germ cell development and sperm functions as well. This review aims to summarize recent findings regarding kisspeptin activity in the testes and sperm function. METHODS We undertook a keyword search of peer-reviewed research literature including data from in vivo and in vitro studies in humans and genetically modified animal models to identify the roles of kisspeptins in male reproduction. RESULTS A plethora of studies detail the role of kisspeptins and GPR54 in mammalian spermatogenesis in vivo and in vitro. This review identified recent findings regarding the kisspeptin system in male gonads, and regulation of kisspeptin in testicular physiology and male reproductive defects and disorders. CONCLUSION The findings of this review confirm the importance role of kisspeptins in male fertility. Understanding their biphasic roles in testis may help to consider kisspeptins as potential pharmacological targets for treating human infertility.
Collapse
Affiliation(s)
- Tao Feng
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, 100097, China
| | - Jia H Bai
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, 100097, China
| | - Xiao L Xu
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, 100097, China
| | - Yan Liu
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, 100097, China
| |
Collapse
|
78
|
Wang XQ, Fang PF, Zhang C, Xu YY, Song XB, Liang J, Xia QR. Low KISS1 expression predicts poor prognosis for patients with colorectal cancer: A meta-analysis. Clin Exp Pharmacol Physiol 2019; 46:625-634. [PMID: 30932210 DOI: 10.1111/1440-1681.13093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 03/22/2019] [Accepted: 03/26/2019] [Indexed: 12/24/2022]
Abstract
KISS1 and KISS1R, a novel pair of metastasis suppressors, are likely to be associated with the prognosis of colorectal cancer (CRC). Here, a meta-analysis was performed to study the role of KISS1 and KISS1R in CRC. Heterogeneity, stability and publication bias were all estimated. Six publications describing a total of 559 CRC patients were included in the present study. Low KISS1 expression predicted 70% higher risk of poor prognosis for general patients (HR, 1.71; 95% CI, 1.28-2.29) and 99% higher risk for East Asian patients (HR, 1.99; 95% CI, 1.46-2.72). Limited evidence indicated that decreased KISS1R expression might predict poor outcome (HR, 2.96; 95% CI, 1.51-5.82). Neither heterogeneity nor publication bias was identified. The current analyses suggest that low KISS1 expression predicts poor overall survival among East Asian patients with CRC. Evidence on other races and KISS1R are still insufficient, and additional studies are required to clarify the risk of CRC associated with KISS1R by race.
Collapse
Affiliation(s)
| | | | - Cheng Zhang
- Anhui Provincial Cancer Institute, Hefei, China.,The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ya-Yun Xu
- Department of Pharmacy, Hefei Fourth People's Hospital, Hefei, China.,Anhui Mental Health Center, Hefei, China
| | | | - Jun Liang
- Department of Pharmacy, Hefei Fourth People's Hospital, Hefei, China.,Anhui Mental Health Center, Hefei, China
| | - Qing-Rong Xia
- Department of Pharmacy, Hefei Fourth People's Hospital, Hefei, China.,Anhui Mental Health Center, Hefei, China
| |
Collapse
|
79
|
Association of Kiss1 and GPR54 Gene Polymorphisms with Polycystic Ovary Syndrome among Sri Lankan Women. BIOMED RESEARCH INTERNATIONAL 2019; 2019:6235680. [PMID: 30993114 PMCID: PMC6434290 DOI: 10.1155/2019/6235680] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 02/21/2019] [Accepted: 02/25/2019] [Indexed: 01/07/2023]
Abstract
Polycystic ovary syndrome (PCOS) is the commonest endocrine disorder affecting women of reproductive age. Its aetiology, though yet unclear, is presumed to have an oligogenic basis interacting with environmental factors. Kisspeptins are peptide products of Kiss1 gene that control the hypothalamic pituitary (HPG) axis by acting via G protein-coupled receptor known as GPR54. There is paucity of data on the role of Kiss1 and GPR54 gene in PCOS. We aimed to identify the polymorphisms in Kiss1 and GPR54 genes and explore their association with serum kisspeptin levels among Sri Lankan women with well-characterized PCOS. Consecutive women with PCOS manifesting from adolescence (n=55) and adult controls (n=110) were recruited. Serum kisspeptin and testosterone levels were determined by ELISA method. Whole gene sequencing was performed to identify the polymorphisms in Kiss1 and GPR54 genes. Serum kisspeptin and testosterone concentrations were significantly higher in women with PCOS than controls: kisspeptin 4.873nmol/L versus 4.127nmol/L; testosterone 4.713nmol/L versus 3.415 nmol/L, p<0.05. Sequencing the GPR54 gene revealed 5 single nucleotide polymorphisms (SNPs), rs10407968, rs1250729403, rs350131, chr19:918686, and chr19:918735, with two novel SNPs (chr19:918686 and chr19:918735), while sequencing the Kiss1 gene revealed 2 SNPs, rs5780218 and rs4889. All identified SNPs showed no significant difference in frequency between patients and controls. GPR54 gene rs350131 polymorphism (G/T) was detected more frequently in our study population. The heterozygous allele (AG) of GPR54 gene novel polymorphism chr19:918686 showed a marginal association with serum kisspeptin levels (p=0.053). Genetic variations in GPR54 and Kiss1 genes are unlikely to be associated with PCOS among Sri Lankan women manifesting from adolescence. Meanwhile the heterozygous allele of chr19:918686 is probably associated with serum kisspeptin concentrations, which suggests a potential role in the aetiology of PCOS.
Collapse
|
80
|
Aquino NSS, Kokay IC, Perez CT, Ladyman SR, Henriques PC, Silva JF, Broberger C, Grattan DR, Szawka RE. Kisspeptin Stimulation of Prolactin Secretion Requires Kiss1 Receptor but Not in Tuberoinfundibular Dopaminergic Neurons. Endocrinology 2019; 160:522-533. [PMID: 30668693 DOI: 10.1210/en.2018-00932] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 01/14/2019] [Indexed: 02/07/2023]
Abstract
Kisspeptin has been shown to stimulate prolactin secretion. We investigated whether kisspeptin acts through the Kiss1 receptor (Kiss1r) to regulate dopamine and prolactin. Initially, we evaluated prolactin response in a Kiss1r-deficient mouse line, in which Kiss1r had been knocked into GnRH neurons (Kiss1r-/-R). Intracerebroventricular kisspeptin-10 (Kp-10) increased prolactin release in wild-type but not in Kiss1r-/-R female mice. In ovariectomized, estradiol-treated rats, the Kiss1r antagonist kisspeptin-234 abolished the Kp-10-induced increase in prolactin release but failed to prevent the concomitant reduction in the activity of tuberoinfundibular dopaminergic (TIDA) neurons, as determined by the 3,4-dihydroxyphenylacetic acid/dopamine ratio in the median eminence. Using whole-cell patch clamp recordings in juvenile male rats, we found no direct effect of Kp-10 on the electrical activity of TIDA neurons. In addition, dual-label in situ hybridization in the hypothalamus of female rats showed that Kiss1r is expressed in the periventricular nucleus of the hypothalamus (Pe) and arcuate nucleus of the hypothalamus (ARC) but not in tyrosine hydroxylase (Th)-expressing neurons. Kisspeptin also has affinity for the neuropeptide FF receptor 1 (Npffr1), which was expressed in the majority of Pe dopaminergic neurons but only in a low proportion of TIDA neurons in the ARC. Our findings demonstrate that Kiss1r is necessary to the effect of kisspeptin on prolactin secretion, although TIDA neurons lack Kiss1r and are electrically unresponsive to kisspeptin. Thus, kisspeptin is likely to stimulate prolactin secretion via Kiss1r in nondopaminergic neurons, whereas the colocalization of Npffr1 and Th suggests that Pe dopaminergic neurons may play a role in the kisspeptin-induced inhibition of dopamine release.
Collapse
Affiliation(s)
- Nayara S S Aquino
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ilona C Kokay
- Centre for Neuroendocrinology and Department of Anatomy, University of Otago, Dunedin, New Zealand
| | | | - Sharon R Ladyman
- Centre for Neuroendocrinology and Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Patricia C Henriques
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Juneo F Silva
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Ilhéus, Brazil
| | | | - David R Grattan
- Centre for Neuroendocrinology and Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Raphael E Szawka
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
81
|
A new class of pentapeptide KISS1 receptor agonists with hypothalamic–pituitary–gonadal axis activation. Bioorg Med Chem Lett 2019; 29:654-658. [DOI: 10.1016/j.bmcl.2018.12.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 11/25/2018] [Accepted: 12/08/2018] [Indexed: 01/06/2023]
|
82
|
Comninos AN, Demetriou L, Wall MB, Shah AJ, Clarke SA, Narayanaswamy S, Nesbitt A, Izzi-Engbeaya C, Prague JK, Abbara A, Ratnasabapathy R, Yang L, Salem V, Nijher GM, Jayasena CN, Tanner M, Bassett P, Mehta A, McGonigle J, Rabiner EA, Bloom SR, Dhillo WS. Modulations of human resting brain connectivity by kisspeptin enhance sexual and emotional functions. JCI Insight 2018; 3:121958. [PMID: 30333302 PMCID: PMC6237465 DOI: 10.1172/jci.insight.121958] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 08/17/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Resting brain connectivity is a crucial component of human behavior demonstrated by disruptions in psychosexual and emotional disorders. Kisspeptin, a recently identified critical reproductive hormone, can alter activity in certain brain structures but its effects on resting brain connectivity and networks in humans remain elusive. METHODS We determined the effects of kisspeptin on resting brain connectivity (using functional neuroimaging) and behavior (using psychometric analyses) in healthy men, in a randomized double-blinded 2-way placebo-controlled study. RESULTS Kisspeptin's modulation of the default mode network (DMN) correlated with increased limbic activity in response to sexual stimuli (globus pallidus r = 0.500, P = 0.005; cingulate r = 0.475, P = 0.009). Furthermore, kisspeptin's DMN modulation was greater in men with less reward drive (r = -0.489, P = 0.008) and predicted reduced sexual aversion (r = -0.499, P = 0.006), providing key functional significance. Kisspeptin also enhanced key mood connections including between the amygdala-cingulate, hippocampus-cingulate, and hippocampus-globus pallidus (all P < 0.05). Consistent with this, kisspeptin's enhancement of hippocampus-globus pallidus connectivity predicted increased responses to negative stimuli in limbic structures (including the thalamus and cingulate [all P < 0.01]). CONCLUSION Taken together, our data demonstrate a previously unknown role for kisspeptin in the modulation of functional brain connectivity and networks, integrating these with reproductive hormones and behaviors. Our findings that kisspeptin modulates resting brain connectivity to enhance sexual and emotional processing and decrease sexual aversion, provide foundation for kisspeptin-based therapies for associated disorders of body and mind. FUNDING NIHR, MRC, and Wellcome Trust.
Collapse
Affiliation(s)
- Alexander N Comninos
- Investigative Medicine, Imperial College London, United Kingdom.,Department of Endocrinology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Lysia Demetriou
- Investigative Medicine, Imperial College London, United Kingdom.,Imanova Centre for Imaging Sciences, Imperial College London, United Kingdom
| | - Matthew B Wall
- Imanova Centre for Imaging Sciences, Imperial College London, United Kingdom.,Division of Brain Sciences, Imperial College London, United Kingdom
| | - Amar J Shah
- Investigative Medicine, Imperial College London, United Kingdom
| | - Sophie A Clarke
- Investigative Medicine, Imperial College London, United Kingdom
| | | | | | | | - Julia K Prague
- Investigative Medicine, Imperial College London, United Kingdom
| | - Ali Abbara
- Investigative Medicine, Imperial College London, United Kingdom
| | | | - Lisa Yang
- Investigative Medicine, Imperial College London, United Kingdom
| | - Victoria Salem
- Investigative Medicine, Imperial College London, United Kingdom
| | | | | | - Mark Tanner
- Imanova Centre for Imaging Sciences, Imperial College London, United Kingdom
| | | | - Amrish Mehta
- Department of Neuroradiology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - John McGonigle
- Imanova Centre for Imaging Sciences, Imperial College London, United Kingdom
| | - Eugenii A Rabiner
- Imanova Centre for Imaging Sciences, Imperial College London, United Kingdom.,Centre for Neuroimaging Sciences, King's College London, United Kingdom
| | - Stephen R Bloom
- Investigative Medicine, Imperial College London, United Kingdom
| | - Waljit S Dhillo
- Investigative Medicine, Imperial College London, United Kingdom
| |
Collapse
|
83
|
Abstract
Reproduction is fundamental for the survival of all species and requires meticulous synchronisation of a diverse complement of neural, endocrine and related behaviours. The reproductive hormone kisspeptin (encoded by the KISS1/Kiss1 gene) is now a well-established orchestrator of reproductive hormones, acting upstream of gonadotrophin-releasing hormone (GnRH) at the apex of the hypothalamic–pituitary–gonadal (HPG) reproductive axis. Beyond the hypothalamus, kisspeptin is also expressed in limbic and paralimbic brain regions, which are areas of the neurobiological network implicated in sexual and emotional behaviours. We are now forming a more comprehensive appreciation of extra-hypothalamic kisspeptin signalling and the complex role of kisspeptin as an upstream mediator of reproductive behaviours, including olfactory-driven partner preference, copulatory behaviour, audition, mood and emotion. An increasing body of research from zebrafish to humans has implicated kisspeptin in the integration of reproductive hormones with an overall positive influence on these reproductive behaviours. In this review, we critically appraise the current literature regarding kisspeptin and its control of reproductive behaviour. Collectively, these data significantly enhance our understanding of the integration of reproductive hormones and behaviour and provide the foundation for kisspeptin-based therapies to treat related disorders of body and mind.
Collapse
Affiliation(s)
- Edouard G A Mills
- Section of Endocrinology and Investigative Medicine, Imperial College London, Hammersmith Hospital, London, UK
| | - Waljit S Dhillo
- Section of Endocrinology and Investigative Medicine, Imperial College London, Hammersmith Hospital, London, UK
| | - Alexander N Comninos
- Section of Endocrinology and Investigative Medicine, Imperial College London, Hammersmith Hospital, London, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, UK
| |
Collapse
|
84
|
Harter CJL, Kavanagh GS, Smith JT. The role of kisspeptin neurons in reproduction and metabolism. J Endocrinol 2018; 238:R173-R183. [PMID: 30042117 DOI: 10.1530/joe-18-0108] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Accepted: 06/13/2018] [Indexed: 02/06/2023]
Abstract
Kisspeptin is a neuropeptide with a critical role in the function of the hypothalamic-pituitary-gonadal (HPG) axis. Kisspeptin is produced by two major populations of neurons located in the hypothalamus, the rostral periventricular region of the third ventricle (RP3V) and arcuate nucleus (ARC). These neurons project to and activate gonadotrophin-releasing hormone (GnRH) neurons (acting via the kisspeptin receptor, Kiss1r) in the hypothalamus and stimulate the secretion of GnRH. Gonadal sex steroids stimulate kisspeptin neurons in the RP3V, but inhibit kisspeptin neurons in the ARC, which is the underlying mechanism for positive- and negative feedback respectively, and it is now commonly accepted that the ARC kisspeptin neurons act as the GnRH pulse generator. Due to kisspeptin's profound effect on the HPG axis, a focus of recent research has been on afferent inputs to kisspeptin neurons and one specific area of interest has been energy balance, which is thought to facilitate effects such as suppressing fertility in those with under- or severe over-nutrition. Alternatively, evidence is building for a direct role for kisspeptin in regulating energy balance and metabolism. Kiss1r-knockout (KO) mice exhibit increased adiposity and reduced energy expenditure. Although the mechanisms underlying these observations are currently unknown, Kiss1r is expressed in adipose tissue and potentially brown adipose tissue (BAT) and Kiss1rKO mice exhibit reduced energy expenditure. Recent studies are now looking at the effects of kisspeptin signalling on behaviour, with clinical evidence emerging of kisspeptin affecting sexual behaviour, further investigation of potential neuronal pathways are warranted.
Collapse
Affiliation(s)
- Campbell J L Harter
- School of Human SciencesThe University of Western Australia, Perth, Western Australia, Australia
| | - Georgia S Kavanagh
- School of Human SciencesThe University of Western Australia, Perth, Western Australia, Australia
| | - Jeremy T Smith
- School of Human SciencesThe University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
85
|
Moore AM, Coolen LM, Porter DT, Goodman RL, Lehman MN. KNDy Cells Revisited. Endocrinology 2018; 159:3219-3234. [PMID: 30010844 PMCID: PMC6098225 DOI: 10.1210/en.2018-00389] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 07/05/2018] [Indexed: 12/29/2022]
Abstract
In the past decade since kisspeptin/neurokinin B/dynorphin (KNDy) cells were first identified in the mammalian hypothalamus, a plethora of new research has emerged adding insights into the role of this neuronal population in reproductive neuroendocrine function, including the basis for GnRH pulse generation and the mechanisms underlying the steroid feedback control of GnRH secretion. In this mini-review, we provide an update of evidence regarding the roles of KNDy peptides and their postsynaptic receptors in producing episodic GnRH release and assess the relative contribution of KNDy neurons to the "GnRH pulse generator." In addition, we examine recent work investigating the role of KNDy neurons as mediators of steroid hormone negative feedback and review evidence for their involvement in the preovulatory GnRH/LH surge, taking into account species differences that exist among rodents, ruminants, and primates. Finally, we summarize emerging roles of KNDy neurons in other aspects of reproductive function and in nonreproductive functions and discuss critical unresolved questions in our understanding of KNDy neurobiology.
Collapse
Affiliation(s)
- Aleisha M Moore
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, Mississippi
| | - Lique M Coolen
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, Mississippi
- Department of Physics and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Danielle T Porter
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, Mississippi
| | - Robert L Goodman
- Department of Physiology, Pharmacology, and Neuroscience, West Virginia University, Morgantown, West Virginia
| | - Michael N Lehman
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
86
|
Güvenç M, Aksakal M. Ameliorating effect of kisspeptin-10 on methotrexate-induced sperm damages and testicular oxidative stress in rats. Andrologia 2018; 50:e13057. [PMID: 29862548 DOI: 10.1111/and.13057] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 04/27/2018] [Accepted: 05/02/2018] [Indexed: 12/01/2022] Open
Abstract
The purpose of this study was to determine the kisspeptin-10 (Kiss) administration on the damages in testicular oxidant-antioxidant system, reproductive organ weights and some spermatological characteristics resulted from methotrexate (MTX) exposure. Group 1 (n:6) received saline only; group 2 (n:6) received 50 nmol/kg kisspeptin-10 for 10 days; group 3 (n:10) received single-dose methotrexate 20 mg/kg; and group 4 (n:10) received MTX 20 mg/kg single dose and, after 3 days, received kisspeptin-10, 50 nmol/kg, lasted for 10 days by intraperitoneal injection. At the end of the study, malondialdehyde levels were found to have increased following the application of MTX while showing a significant reduction in group 4 with Kiss administration. With respect to the spermatological parameters, administering MTX decreased motility and increased the rates of abnormal spermatozoa in group 2, while improvements were observed in group 4 in the form of increased motility in the spermatozoa and fewer abnormal spermatozoa. In addition, Kiss treatment provided statistically significant increases in the absolute weight of the seminal vesicles and the relative weights of the right cauda epididymis and seminal vesicles resulting from MTX administration. MTX administration damaged some spermatological parameters and increased oxidative stress when compared to the control group. However, Kiss treatment was observed to mitigate these adverse effects as demonstrated by the improvements in coadministration of Kiss and MTX when compared to the MTX group. It is concluded that Kiss treatment may reduce MTX-induced reproductive toxicity as a potential antioxidant compound.
Collapse
Affiliation(s)
- Mehmet Güvenç
- Department of Physiology, Faculty of Veterinary Medicine, Mustafa Kemal University, Hatay, Turkey
| | - Mesut Aksakal
- Department of Physiology, Faculty of Veterinary Medicine, Firat University, Elazig, Turkey
| |
Collapse
|
87
|
Yang L, Comninos AN, Dhillo WS. Intrinsic links among sex, emotion, and reproduction. Cell Mol Life Sci 2018; 75:2197-2210. [PMID: 29619543 PMCID: PMC5948280 DOI: 10.1007/s00018-018-2802-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 01/27/2018] [Accepted: 03/20/2018] [Indexed: 01/23/2023]
Abstract
Species survival is dependent on successful reproduction. This begins with a desire to mate, followed by selection of a partner, copulation and in monogamous mammals including humans, requires emotions and behaviours necessary to maintain partner bonds for the benefit of rearing young. Hormones are integral to all of these stages and not only mediate physiological and endocrine processes involved in reproduction, but also act as neuromodulators within limbic brain centres to facilitate the expression of innate emotions and behaviours required for reproduction. A significant body of work is unravelling the roles of several key hormones in the modulation of mood states and sexual behaviours; however, a full understanding of the integration of these intrinsic links among sexual and emotional brain circuits still eludes us. This review summarises the evidence to date and postulates future directions to identify potential psycho-neuroendocrine frameworks linking sexual and emotional brain processes with reproduction.
Collapse
Affiliation(s)
- Lisa Yang
- Section of Endocrinology and Investigative Medicine, Imperial College London, 6th Floor, Commonwealth Building, Hammersmith Hospital Campus, Du Cane Road, London, W12 ONN, UK
| | - Alexander N Comninos
- Section of Endocrinology and Investigative Medicine, Imperial College London, 6th Floor, Commonwealth Building, Hammersmith Hospital Campus, Du Cane Road, London, W12 ONN, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, UK
| | - Waljit S Dhillo
- Section of Endocrinology and Investigative Medicine, Imperial College London, 6th Floor, Commonwealth Building, Hammersmith Hospital Campus, Du Cane Road, London, W12 ONN, UK.
| |
Collapse
|
88
|
Xing R, Liu F, Yang Y, Cui X, Wang T, Xie L, Zhao Y, Fang L, Yi T, Zheng B, Liu M, Chen H. GPR54 deficiency reduces the Treg population and aggravates experimental autoimmune encephalomyelitis in mice. SCIENCE CHINA-LIFE SCIENCES 2018; 61:675-687. [PMID: 29931449 DOI: 10.1007/s11427-017-9269-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 10/17/2017] [Indexed: 12/20/2022]
Abstract
GPR54 is highly expressed in the central nervous system and plays a crucial role in pubertal development. However, GRP54 is also expressed in the immune system, implying possible immunoregulatory functions. Here we investigated the role of GPR54 in T cell and immune tolerance. GPR54 deficiency led to an enlarged thymus, an increased number of thymocytes, and altered thymic micro-architecture starting around puberty, indicating GPR54 function in T-cell development through its regulatory effect on the gonadal system. However, flow cytometry revealed a significant reduction in the peripheral regulatory T cell population and a moderate decrease in CD4 single-positive thymocytes in prepubertal Gpr54-/- mice. These phenotypes were confirmed in chimeric mice with GPR54 deficient bone marrow-derived cells. In addition, we found elevated T cell activation in peripheral and thymic T cells in Gpr54-/- mice. When intact mice were immunized with myelin oligodendrocyte glycoprotein, a more severe experimental autoimmune encephalomyelitis (EAE) developed in the Gpr54-/- mice. Interestingly, aggravated EAE disease was also manifested in castrated and bone marrow chimeric Gpr54-/- mice compared to the respective wild-type control, suggesting a defect in self-tolerance resulting from GPR54 deletion through a mechanism that bypassed sex hormones. These findings demonstrate a novel role for GPR54 in regulating self-tolerant immunity in a sex hormone independent manner.
Collapse
MESH Headings
- Animals
- Disease Susceptibility
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Gene Expression
- Immune Tolerance/immunology
- Lymphocyte Activation/immunology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Myelin-Oligodendrocyte Glycoprotein/administration & dosage
- Myelin-Oligodendrocyte Glycoprotein/toxicity
- Receptors, Kisspeptin-1/deficiency
- Receptors, Kisspeptin-1/genetics
- Receptors, Kisspeptin-1/physiology
- Spleen/immunology
- T-Lymphocyte Subsets/immunology
- T-Lymphocytes, Regulatory/immunology
- Thymus Gland/immunology
Collapse
Affiliation(s)
- Roumei Xing
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Fang Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yiqing Yang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Xueqin Cui
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Tongtong Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Ling Xie
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yongliang Zhao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Lei Fang
- Third Venture Biotechnology Co., Ltd., Nanjing, 210042, China
| | - Tingfang Yi
- Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Houston, Texas, 77030, USA
| | - Biao Zheng
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
- Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Houston, Texas, 77030, USA.
| | - Huaqing Chen
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| |
Collapse
|
89
|
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
|
90
|
Functional examination of novel kisspeptin phosphinic peptides. PLoS One 2018; 13:e0195089. [PMID: 29614094 PMCID: PMC5882139 DOI: 10.1371/journal.pone.0195089] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 03/18/2018] [Indexed: 01/09/2023] Open
Abstract
Kisspeptins acting on their cognate G protein-coupled receptor, kisspeptin receptor, play important roles in the suppression of cancer cell metastasis and regulation of the reproductive system, and therefore are important for therapeutic intervention. All native functional human kisspeptins (kisspeptin-54, kisspsptin-14 and kisspeptin-13) share the 10 amino acids of kisspeptin-10 at their C-terminus (45–54). However, they are inactivated rapidly by matrix metalloproteinases (MMPs) through the cleavage of the peptide bond between glycine51 and leucine52, which limits their clinical applications. Development of MMP-resistant analogues of kisspeptins may provide better therapeutic outputs. In the present study, two kisspeptin phosphinic peptides were designed and synthesized, and their ability to induce phosphorylation of ERK1/2 through kisspeptin receptor and their inhibition on MMP-2 and MMP-9 whose activity correlates with cancer metastasis were assessed. The results showed that one analogue, phosphinic kisspeptin R isomer (PKPR), exhibited kisspeptin receptor-agonistic activity and also inhibitory activity on MMP-2, indicating that PKPR may serve as a lead for the further development of kisspeptin analogues for therapeutic purpose.
Collapse
|
91
|
Franssen D, Tena-Sempere M. The kisspeptin receptor: A key G-protein-coupled receptor in the control of the reproductive axis. Best Pract Res Clin Endocrinol Metab 2018; 32:107-123. [PMID: 29678280 DOI: 10.1016/j.beem.2018.01.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The kisspeptin receptor, Kiss1R, also known as Gpr54, is a G protein-coupled receptor (GPCR), deorphanized in 2001, when it was recognized as canonical receptor for the Kiss1-derived peptides, kisspeptins. In 2003, inactivating mutations of Kiss1R gene were first associated to lack of pubertal maturation and hypogonadotropic hypogonadism in humans and rodents. These seminal findings pointed out the previously unsuspected, essential role of Kiss1R and its ligands in control of reproductive maturation and function. This contention has been fully substantiated during the last decade by a wealth of clinical and experimental data, which has documented a fundamental function of the so-called Kiss1/Kiss1R system in the regulation of puberty onset, gonadotropin secretion and ovulation, as well as the metabolic and environmental modulation of fertility. In this review, we provide a succinct summary of some of the most salient facets of Kiss1R, as essential GPCR for the proper maturation and function of the reproductive axis.
Collapse
Affiliation(s)
- Delphine Franssen
- Instituto Maimónides de Investigación Biomédica de Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004, Cordoba, Spain; Hospital Universitario Reina Sofia, 14004, Cordoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 14004, Cordoba, Spain
| | - Manuel Tena-Sempere
- Instituto Maimónides de Investigación Biomédica de Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004, Cordoba, Spain; Hospital Universitario Reina Sofia, 14004, Cordoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 14004, Cordoba, Spain; FiDiPro Program, Institute of Biomedicine, University of Turku, FIN-20520, Turku, Finland.
| |
Collapse
|
92
|
Altered aspects of anxiety-related behavior in kisspeptin receptor-deleted male mice. Sci Rep 2018; 8:2794. [PMID: 29434234 PMCID: PMC5809376 DOI: 10.1038/s41598-018-21042-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 01/29/2018] [Indexed: 12/24/2022] Open
Abstract
The roles of kisspeptin signaling outside the hypothalamus in the brain are unknown. We examined here the impact of Kiss1r-deletion on hippocampus-related behaviors of anxiety and spatial learning in adult male mice using two mouse models. In the first, global Kiss1r-null and control mice were gonadectomized (GDX KISS1R-KO). In the second, KISS1R signalling was rescued selectively in gonadotropin-releasing hormone neurons to generate Kiss1r-null mice with normal testosterone levels (intact KISS1R-KO). Intact KISS1R-KO rescue mice were found to spend twice as much time in the open arms of the elevated plus maze (EPM) compared to controls (P < 0.01). GDX KISS1R-KO mice showed a similar but less pronounced trend. No differences were detected between intact KISS1R-KO mice and controls in the open field test (OFT), although a marked reduction in time spent in the centre quadrant was observed for all GDX mice (P < 0.001). No effects of KISS1R deletion or gonadectomy were detected in the Morris water maze. These observations demonstrate that KISS1R signalling impacts upon anxiogenic neural circuits operative in the EPM, while gonadal steroids appear important for anxiety behaviour observed in the OFT. The potential anxiogenic role of kisspeptin may need to be considered in the development of kisspeptin analogs for the clinic.
Collapse
|
93
|
Zhu B, Ge W. Genome editing in fishes and their applications. Gen Comp Endocrinol 2018; 257:3-12. [PMID: 28919449 DOI: 10.1016/j.ygcen.2017.09.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Revised: 08/15/2017] [Accepted: 09/13/2017] [Indexed: 12/18/2022]
Abstract
There have been revolutionary progresses in genome engineering in the past few years. The newly-emerged genome editing technologies including zinc-finger nuclease (ZFN), transcription activator-like effector nuclease (TALEN) and clustered regularly interspaced short palindromic repeats associated with Cas9 (CRISPR/Cas9) have enabled biological scientists to perform efficient and precise targeted genome editing in different species. Fish represent the largest group of vertebrates with many species having values for both scientific research and aquaculture industry. Genome editing technologies have found extensive applications in different fish species for basic functional studies as well asapplied research in such fields as disease modeling and aquaculture. This mini-review focuses on recent advancements and applications of the new generation of genome editing technologies in fish species, with particular emphasis on their applications in understanding reproductive functions because the reproductive axis has been most systematically and best studied among others and its function has been difficult to address with reverse genetics approach.
Collapse
Affiliation(s)
- Bo Zhu
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Wei Ge
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau, China.
| |
Collapse
|
94
|
Chianese R, Colledge WH, Fasano S, Meccariello R. Editorial: The Multiple Facets of Kisspeptin Activity in Biological Systems. Front Endocrinol (Lausanne) 2018; 9:727. [PMID: 30559719 PMCID: PMC6286967 DOI: 10.3389/fendo.2018.00727] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 11/16/2018] [Indexed: 11/21/2022] Open
Affiliation(s)
- Rosanna Chianese
- Department of Experimental Medicine, University of Campania L. Vanvitelli, Naples, Italy
| | - William H. Colledge
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Silvia Fasano
- Department of Experimental Medicine, University of Campania L. Vanvitelli, Naples, Italy
| | - Rosaria Meccariello
- Department of Movement Sciences and Wellbeing, Parthenope University of Naples, Naples, Italy
- *Correspondence: Rosaria Meccariello
| |
Collapse
|
95
|
Wahab F, Atika B, Ullah F, Shahab M, Behr R. Metabolic Impact on the Hypothalamic Kisspeptin-Kiss1r Signaling Pathway. Front Endocrinol (Lausanne) 2018; 9:123. [PMID: 29643834 PMCID: PMC5882778 DOI: 10.3389/fendo.2018.00123] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 03/12/2018] [Indexed: 12/12/2022] Open
Abstract
A large body of data has established the hypothalamic kisspeptin (KP) and its receptor, KISS1R, as major players in the activation of the neuroendocrine reproductive axis at the time of puberty and maintenance of reproductive capacity in the adult. Due to its strategic location, this ligand-receptor pair acts as an integrator of cues from gonadal steroids as well as of circadian and seasonal variation-related information on the reproductive axis. Besides these cues, the activity of the hypothalamic KP signaling is very sensitive to the current metabolic status of the body. In conditions of energy imbalance, either positive or negative, a number of alterations in the hypothalamic KP signaling pathway have been documented in different mammalian models including nonhuman primates and human. Deficiency of metabolic fuels during fasting causes a marked reduction of Kiss1 gene transcript levels in the hypothalamus and, hence, decreases the output of KP-containing neurons. Food intake or exogenous supply of metabolic cues, such as leptin, reverses metabolic insufficiency-related changes in the hypothalamic KP signaling. Likewise, alterations in Kiss1 expression have also been reported in other situations of energy imbalance like diabetes and obesity. Information related to the body's current metabolic status reaches to KP neurons both directly as well as indirectly via a complex network of other neurons. In this review article, we have provided an updated summary of the available literature on the regulation of the hypothalamic KP-Kiss1r signaling by metabolic cues. In particular, the potential mechanisms of metabolic impact on the hypothalamic KP-Kiss1r signaling, in light of available evidence, are discussed.
Collapse
Affiliation(s)
- Fazal Wahab
- Platform Degenerative Diseases, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
- *Correspondence: Fazal Wahab,
| | - Bibi Atika
- Department of Developmental Biology, Faculty of Biology, University of Göttingen, Göttingen, Germany
| | - Farhad Ullah
- Department of Zoology, Islamia College University, Peshawar, Pakistan
| | - Muhammad Shahab
- Laboratory of Reproductive Neuroendocrinology, Department of Animal Sciences, Faculty of Biological Sciences, Quiad-i-Azam University, Islamabad, Pakistan
| | - Rüdiger Behr
- Platform Degenerative Diseases, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| |
Collapse
|
96
|
Ogawa S, Parhar IS. Biological Significance of Kisspeptin-Kiss 1 Receptor Signaling in the Habenula of Teleost Species. Front Endocrinol (Lausanne) 2018; 9:222. [PMID: 29867758 PMCID: PMC5949316 DOI: 10.3389/fendo.2018.00222] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 04/19/2018] [Indexed: 12/13/2022] Open
Abstract
Kisspeptin is a neuropeptide, encoded by kisspeptin 1 (KISS1)/Kiss1 gene, which primarily acts as the regulator of reproductive functions via its receptor, kisspeptin receptor (KissR) in vertebrates. In the brain, Kiss1 gene is mainly expressed in the hypothalamic region, but KissR gene is widely distributed throughout the brain, suggesting that kisspeptin-KissR system may be involved in not only reproductive, but also non-reproductive functions. In non-mammalian vertebrates, there are two or more kisspeptin and KissR types. The zebrafish (Danio rerio) possess two kisspeptin (Kiss1 and Kiss2) and their respective receptors [Kiss1 receptor (KissR1) and KissR2]. In the brain of zebrafish, while Kiss2 is expressed in the preoptic-hypothalamic area, Kiss1 is predominantly expressed in the habenula, an evolutionarily conserved epithalamic structure. Similarly, KissR1 is expressed only in the habenula, while KissR2 is widely distributed in the brain, suggesting that the two kisspeptin systems play specific roles in the brain. The habenular Kiss1 is involved in the modulation of the raphe nuclei and serotonin-related behaviors such as fear response in the zebrafish. This review summarizes the roles of multiple kisspeptin-KissR systems in reproductive and non-reproductive functions and neuronal mechanism, and debates the biological and evolutional significance of habenular kisspeptin-KissR systems in teleost species.
Collapse
|
97
|
Kasum M, Franulić D, Čehić E, Orešković S, Lila A, Ejubović E. Kisspeptin as a promising oocyte maturation trigger for in vitro fertilisation in humans. Gynecol Endocrinol 2017; 33:583-587. [PMID: 28393578 DOI: 10.1080/09513590.2017.1309019] [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] [Indexed: 12/27/2022] Open
Abstract
The aim of this review is to analyse the effectiveness of exogenous kisspeptin administration as a novel alternative of triggering oocyte maturation, instead of currently used triggers such as human chorionic gonadotropin (hCG) or gonadotropin releasing hormone (GnRH) agonist, in women undergoing in vitro fertilisation (IVF) treatment. Kisspeptin has been considered a master regulator of two modes of GnRH and hence gonadotropin secretion, pulses and surges. Administration of kisspeptin-10 and kisspeptin-54 induces the luteinising hormone (LH) surge required for egg maturation and ovulation in animal investigations and LH release during the preovulatory phase of the menstrual cycle and hypothalamic amenorrhoea in humans. Exogenous kisspeptin-54 has been successfully administered as a promising method of triggering oocyte maturation, following ovarian stimulation with gonadotropins and GnRH antagonists in women undergoing IVF, due to its efficacy considering achieved pregnancy rates compared to hCG and GnRH agonists. Also, its safety in patients at high risk of developing ovarian hyperstimulation syndrome is noteworthy. Nevertheless, further studies would be desirable to establish the optimal trigger of egg maturation and to improve the reproductive outcome for women undergoing IVF treatment.
Collapse
Affiliation(s)
- Miro Kasum
- a Department of Obstetrics and Gynaecology , School of Medicine, University Hospital Centre Zagreb , Zagreb , Croatia
| | - Daniela Franulić
- a Department of Obstetrics and Gynaecology , School of Medicine, University Hospital Centre Zagreb , Zagreb , Croatia
| | - Ermin Čehić
- b Department of Obstetrics and Gynaecology , Cantonal Hospital Zenica , Zenica , Bosnia and Herzegovina , and
| | - Slavko Orešković
- a Department of Obstetrics and Gynaecology , School of Medicine, University Hospital Centre Zagreb , Zagreb , Croatia
| | - Albert Lila
- c Gynaecology Cabinet, Kosovo Occupational Health Institute , Giakove , Kosovo
| | - Emina Ejubović
- b Department of Obstetrics and Gynaecology , Cantonal Hospital Zenica , Zenica , Bosnia and Herzegovina , and
| |
Collapse
|
98
|
Šípková J, Kramáriková I, Hynie S, Klenerová V. The galanin and galanin receptor subtypes, its regulatory role in the biological and pathological functions. Physiol Res 2017; 66:729-740. [PMID: 28730831 DOI: 10.33549/physiolres.933576] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The multitalented neuropeptide galanin was first discovered 30 years ago but initially no biologic activity was found. Further research studies discovered the presence of galanin in the brain and some peripheral tissues, and galanin was identified as a modulator of neurotransmission in the central and peripheral nervous system. Over the last decade there were performed very intensive studies of the neuronal actions and also of nonneuronal actions of galanin. Other galanin family peptides have been described, namely galanin, galanin-like peptide, galanin-message associated peptide and alarin. The effect of these peptides is mediated through three galanin receptors subtypes, GalR1, GalR2 and GalR3 belonging to G protein coupled receptors, and signaling via multiple transduction pathways, including inhibition of cyclic AMP/protein kinase A (GalR1, GalR3) and stimulation of phospholipase C (GalR2). This also explains why one specific molecule of galanin can be responsible for different roles in different tissues. The present review summarizes the information currently available on the relationship between the galaninergic system and known pathological states. The research of novel galanin receptor specific agonists and antagonists is also very promising for its future role in pharmacological treatment. The galaninergic system is important target for current and future biomedical research.
Collapse
Affiliation(s)
- J Šípková
- Laboratory of Neuropharmacology, Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University, Prague, Czech Republic.
| | | | | | | |
Collapse
|
99
|
Zeydabadi Nejad S, Ramezani Tehrani F, Zadeh-Vakili A. The Role of Kisspeptin in Female Reproduction. Int J Endocrinol Metab 2017; 15:e44337. [PMID: 29201072 PMCID: PMC5702467 DOI: 10.5812/ijem.44337] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 02/20/2017] [Accepted: 03/09/2017] [Indexed: 02/05/2023] Open
Abstract
CONTEXT Kisspeptin (KISS1), a recently discovered neuropeptide that acts upstream of gonadotropin-releasing hormone (GnRH) neurons, is critical for maturation and function of the reproductive axis. This review aimed at providing comprehensive and up-to-date information on Kisspeptin and its role in female reproduction. EVIDENCE ACQUISITION A literature review was performed using PubMed for all English language articles published between 1999 and 2016. RESULTS The kisspeptin system (KISS1/G protein-coupled receptor-54,GPR54) has recently been addressed as an essential gatekeeper of puberty onset and gonadotropin secretion. Compelling evidence has documented that hypothalamic Kisspeptin mediates steroid feedback and metabolic cues at different developmental stages throughout lifespan. Furthermore, in pre/postnatally androgenized animal models, which exhibit many of the characteristics of Polycystic Ovarian Syndrome (PCOS), the hypothalamic expression of KISS1 and GnRH is abnormal, which might lead to multiple tissue abnormalities observed in this disorder. CONCLUSIONS Kisspeptin, a principal activator of GnRH neurons and the target of endocrine and metabolic cues, is a prerequisite for the onset of puberty and maintenance of normal reproductive function, as abnormal KISS1/GPR54 system has been reported in both animal models and patients with certain forms of infertility, e.g. Idiopathic Hypogonadotropic hypogonadism (IHH) and PCOS. The information suggests that kisspeptin or its receptor represents a potential therapeutic target in the treatment of patients with fertility disorders.
Collapse
Affiliation(s)
- Sareh Zeydabadi Nejad
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, IR Iran
| | - Fahimeh Ramezani Tehrani
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, IR Iran
| | - Azita Zadeh-Vakili
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, IR Iran
- Corresponding author: Azita Zadeh-Vakili, PhD, Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, IR Iran. Tel: +98-2122432513, Fax: +98-2122402463, E-mail:
| |
Collapse
|
100
|
Aquino NSS, Araujo-Lopes R, Henriques PC, Lopes FEF, Gusmao DO, Coimbra CC, Franci CR, Reis AM, Szawka RE. α-Estrogen and Progesterone Receptors Modulate Kisspeptin Effects on Prolactin: Role in Estradiol-Induced Prolactin Surge in Female Rats. Endocrinology 2017; 158:1812-1826. [PMID: 28387824 DOI: 10.1210/en.2016-1855] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 03/31/2017] [Indexed: 11/19/2022]
Abstract
Kisspeptin (Kp) regulates prolactin (PRL) in an estradiol-dependent manner. We investigated the interaction between ovarian steroid receptors and Kp in the control of PRL secretion. Intracerebroventricular injections of Kp-10 or Kp-234 were performed in ovariectomized (OVX) rats under different hormonal treatments. Kp-10 increased PRL release and decreased 3,4-dihydroxyphenylacetic acid levels in the median eminence (ME) of OVX rats treated with estradiol (OVX+E), which was prevented by tamoxifen. Whereas these effects of Kp-10 were absent in OVX rats, they were replicated in OVX rats treated with selective agonist of estrogen receptor (ER)α, propylpyrazole triol, but not of ERβ, diarylpropionitrile. Furthermore, the Kp-10-induced increase in PRL was two times higher in OVX+E rats also treated with progesterone (OVX+EP), which was associated with a reduced expression of both tyrosine hydroxylase (TH) and Ser40-phosphorylated TH in the ME. Kp-10 also reduced dopamine levels in the ME of OVX+EP rats, an effect blocked by the progesterone receptor (PR) antagonist RU486. We also determined the effect of Kp antagonism with Kp-234 on the estradiol-induced surges of PRL and luteinizing hormone (LH), using tail-tip blood sampling combined with ultrasensitive enzyme-linked immunosorbent assay. Kp-234 impaired the early phase of the PRL surge and prevented the LH surge in OVX+E rats. Thus, we provide evidence that Kp stimulation of PRL release requires ERα and is potentiated by progesterone via PR activation. Moreover, alongside its essential role in the LH surge, Kp seems to play a role in the peak phase of the estradiol-induced PRL surge.
Collapse
Affiliation(s)
- Nayara S S Aquino
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901, Belo Horizonte, MG, Brazil
| | - Roberta Araujo-Lopes
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901, Belo Horizonte, MG, Brazil
| | - Patricia C Henriques
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901, Belo Horizonte, MG, Brazil
| | - Felipe E F Lopes
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901, Belo Horizonte, MG, Brazil
| | - Daniela O Gusmao
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901, Belo Horizonte, MG, Brazil
| | - Candido C Coimbra
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901, Belo Horizonte, MG, Brazil
| | - Celso R Franci
- Departamento de Fisiologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto 14049-900, São Paulo, Brazil
| | - Adelina M Reis
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901, Belo Horizonte, MG, Brazil
| | - Raphael E Szawka
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901, Belo Horizonte, MG, Brazil
| |
Collapse
|