1
|
Ahern DF, Martins K, Flórez JM, Ross CE, Huisman A, Cushman RA, Shuping SL, Nestor CC, Desaulniers AT, White BR, Sonstegard TS, Lents CA. Development of KISS1 knockout pigs is characterized by hypogonadotropic hypogonadism, normal growth, and reduced skatole†. Biol Reprod 2024; 111:1082-1096. [PMID: 39375014 DOI: 10.1093/biolre/ioae140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/23/2024] [Accepted: 10/03/2024] [Indexed: 10/09/2024] Open
Abstract
Kisspeptin is a major regulator of gonadotropin secretion in pigs. Previously, CRISPR/Cas9 knockout of KISS1 was used to develop a mosaic parental line of pigs to generate offspring that would not need castration due to loss of kisspeptin. The current goal was to characterize growth and reproductive development of F1 pigs from this parental line. Body weights, gonadotropin concentrations and gonadal development were measured from birth through development (boars to 220 days of age, n = 42; gilts to 160 days of age, n = 36). Testosterone, skatole, and androstenone were also measured in boars. Blood samples were collected by jugular venipuncture for quantification of serum hormones, gonadal tissues were collected for gross morphology and histology, and a fat biopsy was collected (boars) for skatole and androstenone analysis. Body weight did not differ with genotype. There were no differences between KISS1+/+ and heterozygote KISS1+/- animals for most parameters measured. Gonadotropin concentrations were reduced in KISS1-/- boars and gilts compared with KISS1+/+ and KISS1+/- animals (P < 0.05). Concentrations of testosterone in serum and both androstenone and skatole in adipose were less in KISS1-/- boars than in KISS1+/+ and KISS1+/- boars (P < 0.05). Hypogonadism was present in all KISS1-/- gilts and boars. These data indicate that knocking out KISS1 causes hypogonadotropic hypogonadism but does not negatively affect growth in pigs. Only one KISS1 allele is needed for normal gonadotropin secretion and gonadal development, and accumulation of compounds in adipose leading to boar taint.
Collapse
Affiliation(s)
- Daniel F Ahern
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE 68583-0908, USA
| | - Kyra Martins
- Acceligen Inc., 3388 Mike Collins Drive, Eagan, MN 55121, USA
| | - Julio M Flórez
- Acceligen Inc., 3388 Mike Collins Drive, Eagan, MN 55121, USA
- Department of Preventive Veterinary Medicine and Animal Reproduction, School of Agricultural and Veterinarian Sciences, São Paulo State University (Unesp), 14884-900 Via de Acesso Prof. Paulo Donato Castellane s/n, Jaboticabal, Brazil
| | - Caitlin E Ross
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE 68583-0908, USA
| | - Abe Huisman
- Hypor, Hendrix Genetics, Villa `de Körver', Spoorstraat 69, 5831 CK Boxmeer, Netherlands
| | - Robert A Cushman
- United States Department of Agriculture (USDA), Agriculture Research Service (ARS), U.S. Meat Animal Research Center, Livestock Bio-systems Research Unit, Clay Center, NE 68933-0165, USA
| | - Sydney L Shuping
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695-7621, USA
| | - Casey C Nestor
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695-7621, USA
| | - Amy T Desaulniers
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583-0905, USA
| | - Brett R White
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE 68583-0908, USA
| | | | - Clay A Lents
- United States Department of Agriculture (USDA), Agriculture Research Service (ARS), U.S. Meat Animal Research Center, Livestock Bio-systems Research Unit, Clay Center, NE 68933-0165, USA
| |
Collapse
|
2
|
Joy KP, Chaube R. Kisspeptin control of hypothalamus-pituitary-ovarian functions. VITAMINS AND HORMONES 2024; 127:153-206. [PMID: 39864941 DOI: 10.1016/bs.vh.2024.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
The discovery of Kisspeptin (Kiss) has opened a new direction in research on neuroendocrine control of reproduction in vertebrates. Belonging to the RF amide family of peptides, Kiss and its cognate receptor Gpr54 (Kissr) have a long and complex evolutionary history. Multiple forms of Kiss and Kissr are identified in non-mammalian vertebrates, with the exception of birds, and monotreme mammals. However, only a single form of the ligand (KISS1/Kiss1) and receptor (KISS1R/Kiss1r) is retained in higher mammals. Kiss1 is distributed in the hypothalamus-pituitary-gonadal (HPG) axis and its primary function is to stimulate gonadotropin-releasing hormone (GnRH) secretion. Kiss1 neurons are distributed in the rostral periventricular area of the third ventricle (RP3V) and arcuate/infundibular nucleus (ARN/IFN). The ARN/IFN is considered the GnRH pulse generator controlled by steroid negative feedback, and the RP3V neurons is concerned with GnRH surge induced by steroid positive feedback in females. The Kiss1-Kiss1r signaling is important in all aspects of reproduction: puberty onset, maintenance of adult gonadal functions and reproductive aging, and hence assumes therapeutic potentials in the treatment of reproductive dysfunctions and induction of artificial reproduction. This chapter reviews involvement of Kiss1 in the control of the HPG axis functions in female mammals.
Collapse
Affiliation(s)
- K P Joy
- Retired Professor, Department of Zoology, Banaras Hindu University, Varanasi, Uttar pradesh, India.
| | - R Chaube
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar pradesh, India
| |
Collapse
|
3
|
Chou EL, Lu E, Dake MD, Fischbein MP, Bavaria JE, Oderich G, Makaroun MS, Charlton-Ouw KM, Naslund T, Suckow BD, Matsumura JS, Patel HJ, Azizzadeh A. Initial Outcomes of the Gore TAG Thoracic Branch Endoprosthesis for Endovascular Repair of Blunt Thoracic Aortic Injury. Ann Vasc Surg 2024; 104:147-155. [PMID: 38492730 DOI: 10.1016/j.avsg.2023.12.088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 03/18/2024]
Abstract
BACKGROUND Endovascular repair of blunt thoracic aortic injury (BTAI) has dramatically reduced the morbidity and mortality of intervention. Injuries requiring zone 2 coverage of the aorta traditionally require left subclavian artery (LSA) sacrifice or open revascularization. Furthermore, these injuries are associated with an increased risk of in-hospital mortality and long-term morbidity. Here we report 1-year outcomes of total endovascular repair of BTAI with the GORE® TAG® Thoracic Branch Endoprosthesis for LSA preservation. METHODS Across 34 investigative sites, 9 patients with BTAI requiring LSA coverage were enrolled in a nonrandomized, prospective study of a single-branched aortic endograft. The thoracic branch endoprosthesis device allows for graft placement proximal to the LSA and incorporates a single side branch for LSA perfusion. RESULTS This initial cohort included 8 male and 1 female patient with a median age of 43 (22, 76) and 12 months of follow-up. Five total years of follow-up are planned. All participants had grade 3 BTAI. All procedures took place between 2018 and 2019. The median injury severity score was 2 (0, 66). The median procedure time was 109 min (78, 162). All aortic injuries were repaired under general anesthesia and with heparinization. A spinal drain was used in one patient. Post-deployment balloon angioplasty was conducted in one case at the distal landing zone. There was one asymptomatic LSA branch occlusion 6 months after repair. It was attributed to the purposeful proximal deployment of the branch stent to accommodate an early vertebral takeoff. The occlusion did not require revascularization. There were no strokes, mortalities, or aortic adverse events (migration, endoleak, native aortic expansion, dissection, or thrombosis) through 12 months of follow-up. CONCLUSIONS Initial cohort outcomes suggest that endovascular repair of zone 2 BTAI is feasible and has favorable outcomes using the thoracic branch device with LSA preservation. Additional cases and longer-term follow-up are required for a definitive assessment of the device's safety and durability in traumatic aortic injuries.
Collapse
Affiliation(s)
- Elizabeth L Chou
- Division of Vascular Surgery, Cedars-Sinai Medical Center, Los Angeles, CA.
| | - Eileen Lu
- Division of Vascular Surgery, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Michael D Dake
- Department of Medical Imaging, University of Arizona Health System, Tucson, AZ
| | - Michael P Fischbein
- Department of Cardiothoracic Surgery, Stanford University Hospitals, Palo Alto, CA
| | - Joseph E Bavaria
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PE
| | | | - Michel S Makaroun
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PE
| | - Kristofer M Charlton-Ouw
- Department of Cardiothoracic and Vascular Surgery, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX
| | - Thomas Naslund
- Department of Vascular Surgery, Vanderbilt University Medical Center, Nashville, TN
| | - Bjoern D Suckow
- Department of Surgery, Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Jon S Matsumura
- Division of Vascular Surgery, Department of Surgery, University of Colorado School of Medicine, Aurora, CO
| | - Himanshu J Patel
- Department of Cardiac Surgery, University of Michigan Frankel Cardiovascular Center, Ann Arbor, MI
| | - Ali Azizzadeh
- Division of Vascular Surgery, Cedars-Sinai Medical Center, Los Angeles, CA
| |
Collapse
|
4
|
McGrath BM, Norman ST, Gaspardis CA, Rose JL, Scott CJ. Characterizing the relationship between gonadotropin releasing hormone (GnRH), kisspeptin, and RFamide related peptide 3 (RFRP-3) neurons in the equine hypothalamus across the estrous cycle and in the anovulatory seasons. Theriogenology 2024; 219:157-166. [PMID: 38432143 DOI: 10.1016/j.theriogenology.2024.02.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 02/26/2024] [Accepted: 02/26/2024] [Indexed: 03/05/2024]
Abstract
To understand better the role that kisspeptin plays in regulating seasonal and estrous cycle changes in the mare, this study investigated the number, location and interactions between GnRH, kisspeptin and RFRP-3 neurons in the equine hypothalamus. Hypothalami were collected from mares during the non-breeding season, vernal transition and various stages of the breeding season. Fluorescent immunohistochemistry was used to label the neuropeptides of interest. GnRH cells were observed primarily in the arcuate nucleus (ARC), while very few labeled cells were identified in the pre-optic area (POA). Kisspeptin cells were identified primarily in the ARC, with a small number of cells observed dorsal to the ARC, surrounding the third ventricle (3V). The mean number of kisspeptin cells varied between animals and typically showed no pattern associated with season or stage of estrous cycle, but a seasonal difference was identified in the ARC population. Small numbers of RFRP-3 cells were observed in the ARC, ventromedial hypothalamus (VMH) and dorsomedial hypothalamus (DMH). The mean number of RFRP-3 cells appeared higher in pre-ovulatory animals compared to all other stages. The percentage of GnRH cell bodies with kisspeptin appositions did not change with season or stage of estrous cycle. The percentage of kisspeptin cells receiving inputs from RFRP-3 fibers did not vary with season or stage of estrous cycle. These interactions suggest the possibility of the presence of an ultra-short loop feedback system between these three peptides. The changes in RFRP-3 neurons suggest the possibility of a role in the regulation of reproduction in the horse, but it is unlikely to be as a gonadotropin inhibitory factor.
Collapse
Affiliation(s)
- B M McGrath
- School of Dentistry & Medical Sciences, Locked bag 588, Charles Sturt University, Wagga Wagga, NSW, 2678, Australia.
| | - S T Norman
- School of Animal and Veterinary Sciences, Locked bag 588, Charles Sturt University, Wagga Wagga, NSW, 2678, Australia.
| | - C A Gaspardis
- School of Animal and Veterinary Sciences, Locked bag 588, Charles Sturt University, Wagga Wagga, NSW, 2678, Australia.
| | - J L Rose
- School of Dentistry & Medical Sciences, Locked bag 588, Charles Sturt University, Wagga Wagga, NSW, 2678, Australia.
| | - C J Scott
- School of Dentistry & Medical Sciences, Locked bag 588, Charles Sturt University, Wagga Wagga, NSW, 2678, Australia.
| |
Collapse
|
5
|
Aerts EG, Griesgraber MJ, Shuping SL, Bowdridge EC, Hardy SL, Goodman RL, Nestor CC, Hileman SM. The effect of NK3-Saporin injection within the arcuate nucleus on puberty, the LH surge, and the response to Senktide in female sheep†. Biol Reprod 2024; 110:275-287. [PMID: 37930247 DOI: 10.1093/biolre/ioad147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/09/2023] [Accepted: 10/24/2023] [Indexed: 11/07/2023] Open
Abstract
The timing of puberty onset is reliant on increased gonadotropin-releasing hormone (GnRH). This elicits a corresponding increase in luteinizing hormone (LH) due to a lessening of sensitivity to the inhibitory actions of estradiol (E2). The mechanisms underlying the increase in GnRH release likely involve a subset of neurons within the arcuate (ARC) nucleus of the hypothalamus that contain kisspeptin, neurokinin B (NKB), and dynorphin (KNDy neurons). We aimed to determine if KNDy neurons in female sheep are critical for: timely puberty onset; the LH surge; and the response to an intravenous injection of the neurokinin-3 receptor (NK3R) agonist, senktide. Prepubertal ewes received injections aimed at the ARC containing blank-saporin (control, n = 5) or NK3-saporin (NK3-SAP, n = 6) to ablate neurons expressing NK3R. Blood samples taken 3/week for 65 days following surgery were assessed for progesterone to determine onset of puberty. Control ewes exhibited onset of puberty at 33.2 ± 3.9 days post sampling initiation, whereas 5/6 NK3-SAP treated ewes didn't display an increase in progesterone. After an artificial LH surge protocol, surge amplitude was lower in NK3-SAP ewes. Finally, ewes were treated with senktide to determine if an LH response was elicited. LH pulses were evident in both groups in the absence of injections, but the response to senktide vs saline was similar between groups. These results show that KNDy cells are necessary for timely puberty onset and for full expresson of the LH surge. The occurrence of LH pulses in NK3-SAP treated ewes may indicate a recovery from an apulsatile state.
Collapse
Affiliation(s)
- Eliana G Aerts
- Department of Physiology, Pharmacology and Toxicology, Morgantown, WV 26506, USA
| | - Max J Griesgraber
- Department of Physiology, Pharmacology and Toxicology, Morgantown, WV 26506, USA
| | - Sydney L Shuping
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA
| | | | - Steven L Hardy
- Department of Physiology, Pharmacology and Toxicology, Morgantown, WV 26506, USA
| | - Robert L Goodman
- Department of Physiology, Pharmacology and Toxicology, Morgantown, WV 26506, USA
- Department of Neuroscience, West Virginia University, Morgantown, WV 26506, USA
| | - Casey C Nestor
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA
| | - Stanley M Hileman
- Department of Physiology, Pharmacology and Toxicology, Morgantown, WV 26506, USA
- Department of Neuroscience, West Virginia University, Morgantown, WV 26506, USA
| |
Collapse
|
6
|
Ludwig M, Newton C, Pieters A, Homer NZM, Feng Li X, O'Byrne KT, Millar RP. Provocative tests with Kisspeptin-10 and GnRH set the scene for determining social status and environmental impacts on reproductive capacity in male African lions (Panthera leo). Gen Comp Endocrinol 2022; 329:114127. [PMID: 36150474 DOI: 10.1016/j.ygcen.2022.114127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 08/25/2022] [Accepted: 09/16/2022] [Indexed: 11/24/2022]
Abstract
Understanding the hypothalamic factors regulating reproduction facilitates maximising the reproductive success of breeding programmes and in the management and conservation of threatened species, including African lions. To provide insight into the physiology and pathophysiology of the hypothalamic-pituitary-gonadal reproductive axis in lions, we studied the luteinising hormone (LH) and steroid hormone responses to gonadotropin-releasing hormone (GnRH) and its upstream regulator, kisspeptin. Six young (13.3 ± 1.7 months, 56.2 ± 4.3 kg) and four adult (40.2 ± 1.4 months, 174 ± 6 kg) male lions (Ukutula Conservation Centre, South Africa) were used in this study. Lions were immobilised with a combination of medetomidine and ketamine and an intravenous catheter was placed in a jugular, cephalic or medial saphenous vein for blood sampling at 10-min intervals for 220 min. The ten-amino acid kisspeptin which has full intrinsic activity (KP-10, 1 µg/kg) and GnRH (1 µg/kg) were administered intravenously to study their effects on LH and steroid hormone plasma concentrations, measured subsequently by ELISA and liquid chromatography tandem mass spectrometry (LC-MS/MS), respectively. Basal LH levels were similarly low between the age groups, but testosterone and its precursor levels were higher in the adult animals. Adult lions showed a significant LH response to KP-10 (10-fold) and GnRH (11-fold) administration (p < 0.05 and P < 0.001, respectively) whereas in young lions LH increased significantly only in response to GnRH. In adults alone, testosterone and its precursors steadily increased in response to KP-10, with no significant further increase in response to GnRH. Plasma levels of glucocorticoids in response to KP-10 remained unchanged. We suggest that provocative testing of LH and steroid stimulation with kisspeptin provides a new and sensitive tool for determining reproductive status and possibly an index of exposure to stress, environmental insults such as disease, endocrine disruptors and nutritional status. 272 words.
Collapse
Affiliation(s)
- Mike Ludwig
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK; Department of Immunology, Centre for Neuroendocrinology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa.
| | - Claire Newton
- Department of Immunology, Centre for Neuroendocrinology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Ané Pieters
- Department of Immunology, Centre for Neuroendocrinology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Natalie Z M Homer
- BHF/University Centre for Cardiovascular Science and Mass Spectrometry Core, University of Edinburgh, UK
| | - Xiao Feng Li
- Department of Women and Children's Health, School of Life Course Sciences, King's College London, London, UK
| | - Kevin T O'Byrne
- Department of Women and Children's Health, School of Life Course Sciences, King's College London, London, UK
| | - Robert P Millar
- Department of Immunology, Centre for Neuroendocrinology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa.
| |
Collapse
|
7
|
Bailey VN, Sones JL, Camp CM, Gomes VC, Oberhaus EL. Endocrine and ovarian responses to combined estradiol benzoate-sulpiride in seasonally anovulatory mares treated with kisspeptin. Anim Reprod Sci 2022; 247:107087. [DOI: 10.1016/j.anireprosci.2022.107087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 11/28/2022]
|
8
|
Fanelli D, Beltramo M, Conte G, Cerretini B, Lomet D, Rota A, Aucagne V, Camillo F, Panzani D. The Kisspeptin analogue C6 induces ovulation in jennies. Theriogenology 2022; 189:107-112. [PMID: 35738032 DOI: 10.1016/j.theriogenology.2022.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 06/03/2022] [Accepted: 06/12/2022] [Indexed: 10/18/2022]
Abstract
Kisspeptins (KPs) are the most potent stimulating neurotransmitters of GnRH release, and consequently KP administration triggers LH and/or FSH release. In small ruminants, KP or its analogs induced an LH surge followed by ovulation in both cyclic and acyclic animals, while in the mare KP only increased LH plasma levels but failed to induce ovulation. This study in jennies compares the endocrinological effects, ovulatory and pregnancy rates of the KP analog C6 and the GnRH analog buserelin acetate. The ovarian activity of nine Amiata jennies was monitored daily by transrectal ultrasound for three complete estrous cycles. Jennies in estrus were assigned, to one of three treatment groups: 50 nmol of the KP analog C6 (injected twice, 24 h apart, C6 group); 0.4 mg buserelin acetate (injected once, Bu group); and 2 mL of saline (injected once, CTRL group). Blood samples were collected at Day-1 (-24 h) Day0 (h0, before treatment), h2, h4, h6, h8, h10, h24 (before second treatment with C6), h26, h28, h30, h32, h34, h48 and every 24 h until ovulation. Jennies were inseminated once at h24 with fresh extended semen from a donkey stallion. Pregnancy diagnoses were performed 14 days after ovulation. On days 5, 10, and 14 after ovulation, for every CL the cross-sectional area (CSA) and the vascularized area (VA) were recorded by color doppler ultrasound and measured. Significantly higher plasma LH levels were found after induction between the Bu and CTRL groups at h6 and h8 (P < 0.05), while tendentially higher differences were found between the Bu/C6 groups and CTRL at h10. Five/9, 4/9, and 2/9 jennies ovulated between 24 and 48 h after induction from the Bu, C6, and CTRL groups respectively, (P > 0.05). Correlations between corpora lutea CSA and VA with serum progesterone concentration were r = 0.31, P = 0.01, r = 0.38, P = 0.01, respectively. Pregnancy rates after artificial insemination did not differ among groups (CTRL: 6/9, 66.7%; C6: 7/9, 77.8%; Bu: 6/9, 66.7%; P > 0.05). Ovulation rates after C6 treatment were comparable to that of Bu, although not different from the CTRL. Pregnancy rates were comparable to the literature in terms of fresh extended donkey semen in every group. This study suggests that stimulation of the Kp system in jennies, in contrast to findings observed in mares, induces ovulation. Further studies using higher doses and/or more animals are needed to better characterize the efficacy of C6 in jennies.
Collapse
Affiliation(s)
- Diana Fanelli
- Ospedale Didattico Veterinario "Mario Modenato", Dipartimento di Scienze Veterinarie, Università di Pisa. Via Livornese (Lato Monte), 1289, 56122, San Piero a Grado, Pisa, PI, Italy
| | - Massimiliano Beltramo
- Physiologie de la Reproduction et des Comportements (PR China) UMR7247 CNRS, IFCE, INRAE, Université de Tours, F-37380, Nouzilly, France
| | - Giuseppe Conte
- Dipartimento di Scienze Agrarie, Alimentari e Agro-ambientali, University of Pisa, Pisa, Italy
| | - Benedetta Cerretini
- Ospedale Didattico Veterinario "Mario Modenato", Dipartimento di Scienze Veterinarie, Università di Pisa. Via Livornese (Lato Monte), 1289, 56122, San Piero a Grado, Pisa, PI, Italy
| | - Didier Lomet
- Physiologie de la Reproduction et des Comportements (PR China) UMR7247 CNRS, IFCE, INRAE, Université de Tours, F-37380, Nouzilly, France
| | - Alessandra Rota
- Ospedale Didattico Veterinario "Mario Modenato", Dipartimento di Scienze Veterinarie, Università di Pisa. Via Livornese (Lato Monte), 1289, 56122, San Piero a Grado, Pisa, PI, Italy
| | - Vincent Aucagne
- Center for Molecular Biophysics, CNRS, Rue Charles Sadron CS 80054 45071 ORLEANS Cedex 2, Orleans, France
| | - Francesco Camillo
- Ospedale Didattico Veterinario "Mario Modenato", Dipartimento di Scienze Veterinarie, Università di Pisa. Via Livornese (Lato Monte), 1289, 56122, San Piero a Grado, Pisa, PI, Italy
| | - Duccio Panzani
- Ospedale Didattico Veterinario "Mario Modenato", Dipartimento di Scienze Veterinarie, Università di Pisa. Via Livornese (Lato Monte), 1289, 56122, San Piero a Grado, Pisa, PI, Italy.
| |
Collapse
|
9
|
Salzano G, Robert V, Lomet D, Decourt C, Hommet E, Derouin-Tochon F, Hellier V, Savina F, Vishwanatha TM, Aucagne V, Gref R, Beltramo M. A customized long acting formulation of the kisspeptin analog C6 triggers ovulation in anestrus ewe. J Neuroendocrinol 2022; 34:e13121. [PMID: 35355344 DOI: 10.1111/jne.13121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 03/07/2022] [Accepted: 03/10/2022] [Indexed: 11/30/2022]
Abstract
The modulation of the kisspeptin system holds promise as a treatment for human reproductive disorders and for managing livestock breeding. The design of analogs has overcome some unfavorable properties of the endogenous ligands. However, for applications requiring a prolongation of drug activity, such as ovulation induction in the ewe during the non-breeding season, additional improvement is required. To this aim, we designed and tested three formulations containing the kisspeptin analog C6. Two were based on polymeric nanoparticles (NP1 and NP2) and the third was based on hydrogels composed of a mixture of cyclodextrin polymers and dextran grafted with alkyl side chains (MD/pCD). Only the MD/pCD formulation prolonged C6 activity, as shown by monitoring luteinizing hormone (LH) plasma concentration (elevation duration 23.4 ± 6.1, 13.7 ± 4.7 and 12.0 ± 2.4 h for MD/pCD, NP1 and NP2, respectively). When compared with the free C6 (15 nmol/ewe), the formulated (MD/pCD) doses of 10, 15 and 30 nmol/ewe, but not the 90 nmol/ewe dose, provided a more gradual release of C6 as shown by an attenuated LH release during the first 6 h post-treatment. When tested during the non-breeding season without progestogen priming, only, the formulated 30 nmol/ewe dose triggered ovulation (50% of ewes). Hence, we showed that a formulation with an adapted action time would improve the efficacy of C6 with respect to inducing ovulation during the non-breeding season. This result suggests that formulations containing a kisspeptin analog might find applications in the management of livestock reproduction but also point to the possibility of their use for the treatment of some human reproductive pathologies.
Collapse
Affiliation(s)
- Giuseppina Salzano
- Institut des Sciences Moléculaires d'Orsay (CNRS UMR 8214), Université Paris Saclay, Orsay, France
| | - Vincent Robert
- Physiologie de la Reproduction et des Comportements (PRC) UMR7247 CNRS, IFCE, INRAE, Université de Tours, Nouzilly, France
| | - Didier Lomet
- Physiologie de la Reproduction et des Comportements (PRC) UMR7247 CNRS, IFCE, INRAE, Université de Tours, Nouzilly, France
| | - Caroline Decourt
- Physiologie de la Reproduction et des Comportements (PRC) UMR7247 CNRS, IFCE, INRAE, Université de Tours, Nouzilly, France
| | - Elise Hommet
- Physiologie de la Reproduction et des Comportements (PRC) UMR7247 CNRS, IFCE, INRAE, Université de Tours, Nouzilly, France
| | - Flavie Derouin-Tochon
- Physiologie de la Reproduction et des Comportements (PRC) UMR7247 CNRS, IFCE, INRAE, Université de Tours, Nouzilly, France
| | - Vincent Hellier
- Physiologie de la Reproduction et des Comportements (PRC) UMR7247 CNRS, IFCE, INRAE, Université de Tours, Nouzilly, France
| | - Farah Savina
- Institut des Sciences Moléculaires d'Orsay (CNRS UMR 8214), Université Paris Saclay, Orsay, France
| | | | - Vincent Aucagne
- Centre de Biophysique Moléculaire (CNRS UPR 4301), Orléans, France
| | - Ruxandra Gref
- Institut des Sciences Moléculaires d'Orsay (CNRS UMR 8214), Université Paris Saclay, Orsay, France
| | - Massimiliano Beltramo
- Physiologie de la Reproduction et des Comportements (PRC) UMR7247 CNRS, IFCE, INRAE, Université de Tours, Nouzilly, France
| |
Collapse
|
10
|
Ikegami K, Watanabe Y, Nakamura S, Goto T, Inoue N, Uenoyama Y, Tsukamura H. Cellular and molecular mechanisms regulating the KNDy neuronal activities to generate and modulate GnRH pulse in mammals. Front Neuroendocrinol 2022; 64:100968. [PMID: 34808231 DOI: 10.1016/j.yfrne.2021.100968] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/18/2021] [Accepted: 11/15/2021] [Indexed: 12/30/2022]
Abstract
Accumulating findings during the past decades have demonstrated that the hypothalamic arcuate kisspeptin neurons are supposed to be responsible for pulsatile release of gonadotropin-releasing hormone (GnRH) to regulate gametogenesis and steroidogenesis in mammals. The arcuate kisspeptin neurons express neurokinin B (NKB) and dynorphin A (Dyn), thus, the neurons are also referred to as KNDy neurons. In the present article, we mainly focus on the cellular and molecular mechanisms underlying GnRH pulse generation, that is focused on the action of NKB and Dyn and an interaction between KNDy neurons and astrocytes to control GnRH pulse generation. Then, we also discuss the factors that modulate the activity of KNDy neurons and consequent pulsatile GnRH/LH release in mammals.
Collapse
Affiliation(s)
- Kana Ikegami
- Graduate School of Science, The University of Tokyo, Tokyo 113-0033, Japan
| | - Youki Watanabe
- Graduate School of Applied Life Science, Nippon Veterinary and Life Science University, Tokyo 180-8602, Japan
| | - Sho Nakamura
- Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Ehime 794-8555, Japan
| | - Teppei Goto
- RIKEN Center for Biosystems Dynamics Research, Hyogo 650-0047, Japan
| | - Naoko Inoue
- Graduate School of Bioagricultural Science, Nagoya University, Nagoya 464-8601, Japan
| | - Yoshihisa Uenoyama
- Graduate School of Bioagricultural Science, Nagoya University, Nagoya 464-8601, Japan
| | - Hiroko Tsukamura
- Graduate School of Bioagricultural Science, Nagoya University, Nagoya 464-8601, Japan.
| |
Collapse
|
11
|
Hypothalamic kisspeptin and kisspeptin receptors: Species variation in reproduction and reproductive behaviours. Front Neuroendocrinol 2022; 64:100951. [PMID: 34757093 DOI: 10.1016/j.yfrne.2021.100951] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/22/2021] [Accepted: 10/26/2021] [Indexed: 02/07/2023]
Abstract
Kisspeptin, encoded by the KISS1 gene, was first discovered as a potential metastasis suppressor gene. The prepro-kisspeptin precursor is cleaved into shorter mature bioactive peptides of varying sizes that bind to the G protein-coupled receptor GPR54 (=KISS1R). Over the last two decades, multiple types of Kiss and KissR genes have been discovered in mammalian and non-mammalian vertebrate species, but they are remarkably absent in birds. Kiss neuronal populations are distributed mainly in the hypothalamus. The KissRs are widely distributed in the brain, including the hypothalamic and non-hypothalamic regions, such as the hippocampus, amygdala, and habenula. The role of KISS1-KISS1R in humans and Kiss1-Kiss1R in rodents is associated with puberty, gonadal maturation, and the reproductive axis. However, recent gene deletion studies in zebrafish and medaka have provided controversial results, suggesting that the reproductive role of kiss is dispensable. This review highlights the evolutionary history, localisation, and significance of Kiss-KissR in reproduction and reproductive behaviours in mammalian and non-mammalian vertebrates.
Collapse
|
12
|
Zhao S, Guo Z, Xiang W, Wang P. The neuroendocrine pathways and mechanisms for the control of the reproduction in female pigs. Anim Reprod 2021; 18:e20210063. [PMID: 34925558 PMCID: PMC8677349 DOI: 10.1590/1984-3143-ar2021-0063] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 11/10/2021] [Indexed: 01/05/2024] Open
Abstract
Within the hypothalamic-pituitary-gonad (HPG) axis, the major hierarchical component is gonadotropin-releasing hormone (GnRH) neurons, which directly or indirectly receive regulatory inputs from a wide array of regulatory signals and pathways, involving numerous circulating hormones, neuropeptides, and neurotransmitters, and which operate as a final output for the brain control of reproduction. In recent years, there has been an increasing interest in neuropeptides that have the potential to stimulate or inhibit GnRH in the hypothalamus of pigs. Among them, Kisspeptin is a key component in the precise regulation of GnRH neuron secretion activity. Besides, other neuropeptides, including neurokinin B (NKB), neuromedin B (NMB), neuromedin S (NMS), α-melanocyte-stimulating hormone (α-MSH), Phoenixin (PNX), show potential for having a stimulating effect on GnRH neurons. On the contrary, RFamide-related peptide-3 (RFRP-3), endogenous opioid peptides (EOP), neuropeptide Y (NPY), and Galanin (GAL) may play an inhibitory role in the regulation of porcine reproductive nerves and may directly or indirectly regulate GnRH neurons. By combining data from suitable model species and pigs, we aim to provide a comprehensive summary of our current understanding of the neuropeptides acting on GnRH neurons, with a particular focus on their central regulatory pathways and underlying molecular basis.
Collapse
Affiliation(s)
- Shuang Zhao
- College of Bioengineering, Chongqing University, Chongqing, P. R. China
| | - Zongyi Guo
- Chongqing Academy of Animal Sciences, Chongqing, P. R. China
| | - Wei Xiang
- School of Advanced Agriculture and Bioengineering, Yangtze Normal University, Fuling of Chongqing, P. R. China
| | - Pingqing Wang
- College of Bioengineering, Chongqing University, Chongqing, P. R. China
| |
Collapse
|
13
|
Moraes BSS, Curcio BR, Müller V, Bruhn FRP, Santos IPOD, Danielski JNS, Nogueira CEW. Climatic factors affecting gestational length in mares under subtropical climate. J Equine Vet Sci 2021; 107:103751. [PMID: 34802627 DOI: 10.1016/j.jevs.2021.103751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 08/16/2021] [Accepted: 08/20/2021] [Indexed: 11/28/2022]
Abstract
The aim of this study was (i) to evaluate the relationship of climatic factors with gestational length (GL) and (ii) to evaluate the relationship of sire, foal gender and maternal factors with GL in mares. Retrospective data from 470 gestations of 202 respective mares were collected from a Criollo breeding farm in the southern hemisphere. GL was considered as the interval between ovulation and parturition. Climatic and environmental data (temperature, relative humidity, daily sunshine hours, precipitation, temperature humidity index - THI) were obtained daily and the mean values for foaling month and season were calculated. A multiple factor analysis of variance was performed to determine the relationship of the independent variables and interactions related to the mare, foal gender and stallion, and those related to the weather in each season of the year with GL as the dependent variable. The mean ± standard error (SE) of GL for the 470 gestations was 337 ± 0.4 days with a range of 311-363 days. Young-primiparous mares (340±0.9) had longer gestations (P < .001) than young-multiparous (336 ± 0.7) and mature-multiparous (334 ± 0.7) mares. Foal gender had no effect on GL. Gestational length was found to be affected by the stallion. Mares which experienced an autumn and winter gestational period with less daily sunshine hours had an increased GL (P < .05). In conclusion, environmental and climatic factors during pregnancy influenced the GL in mares, specifically, daily sunshine hours, precipitation and THI presented a negative correlation with GL. Young-primiparous Criollo mares presented longer gestational lengths than young or mature multiparous mares.
Collapse
Affiliation(s)
- Bruna S S Moraes
- Department of Veterinary Clinics, Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Bruna R Curcio
- Department of Veterinary Clinics, Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil.
| | - Vitória Müller
- Department of Veterinary Clinics, Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Fabio R P Bruhn
- Department of Preventive Veterinary Medicine, Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Isadora P O D Santos
- Department of Veterinary Clinics, Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Júlio N S Danielski
- Department of Veterinary Clinics, Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Carlos E W Nogueira
- Department of Veterinary Clinics, Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| |
Collapse
|
14
|
Okada CTC, Kaps M, Perez Quesada J, Gautier C, Aurich J, Aurich C. Diestrous Ovulations in Pregnant Mares as a Response to Low Early Postovulatory Progestogen Concentration. Animals (Basel) 2020; 10:ani10122249. [PMID: 33266083 PMCID: PMC7761349 DOI: 10.3390/ani10122249] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 11/25/2020] [Accepted: 11/28/2020] [Indexed: 12/23/2022] Open
Abstract
Simple Summary During early pregnancy in mares, progestogen is synthesized by the primary corpus luteum, which is the only source of progestogen until endometrial cup and accessory corpus luteum formation, from day 36 of pregnancy onwards. In the present study, we investigated the hormonal profile (gonadotrophin and progestogen concentrations) of 11 mares after experimental reduction of primary corpus luteum function. Two pregnancies of each mare were assigned to the control and treatment groups, respectively, and were analyzed until day 34. Low plasma progestogen concentration caused by the treatment reduced the negative feedback on the hypothalamic-pituitary axis, stimulating gonadotropin release, and luteal tissue response. Progestogen concentration restoration soon after treatment suggests a rebound effect and the resurgence of luteal function. In addition, diestrous ovulation was observed between days 11 and 15 in five treatment pregnancies (5/11), but none of the controls (0/11). Although the total luteal area increased after diestrous ovulations, corpus luteum size was not correlated to progestogen secretion. Results suggest that diestrous ovulations during early pregnancy in mares may reflect low progestogen concentrations in the early postovulatory period. Abstract Spontaneous prolongation of the luteal phase has been described in horses, but the underlying causes are still unclear. The present study investigated details of gonadotrophin and progestogen secretion in pregnant mares (n = 11) with or without experimentally reduced early postovulatory luteal function. From days 0 to 3 after ovulation, they were treated with the prostaglandin F2α (PGF2α) analogue cloprostenol or left untreated. After conceptus collection on day 34, they were assigned to the opposite treatment. Mares were affiliated to the group primary corpus luteum (n = 6) or diestrous corpus luteum (n = 5) depending on diestrous corpus luteum (CL) detection in the PGF pregnancy. For statistical comparisons, a p-value < 0.05 was significant. There was an effect of treatment (p < 0.01), but not of group on progestogen concentration. The concentration of LH was higher in PGF-treated than in untreated pregnancies (p < 0.05), but did not differ between groups. The FSH concentration did not differ between groups nor treatments. The total luteal tissue area was greater in mares with a diestrous ovulation during the PGF treatment pregnancy. Low progestogen concentration in the early postovulatory phase diminish the negative feedback on the hypothalamic-pituitary axis in early pregnancy and, thus, stimulate a luteal tissue response. Detection of secondary CL at the time of pregnancy examination in mares may reflect that early post-ovulatory progestogen concentrations were low.
Collapse
Affiliation(s)
- Carolina T. C. Okada
- Platform Artificial Insemination and Embryo Transfer, Department for Small Animals and Horses, Vetmeduni Vienna, Veterinärplatz 1, 1210 Vienna, Austria; (C.T.C.O.); (M.K.); (J.P.Q.); (C.G.)
| | - Martim Kaps
- Platform Artificial Insemination and Embryo Transfer, Department for Small Animals and Horses, Vetmeduni Vienna, Veterinärplatz 1, 1210 Vienna, Austria; (C.T.C.O.); (M.K.); (J.P.Q.); (C.G.)
| | - Javier Perez Quesada
- Platform Artificial Insemination and Embryo Transfer, Department for Small Animals and Horses, Vetmeduni Vienna, Veterinärplatz 1, 1210 Vienna, Austria; (C.T.C.O.); (M.K.); (J.P.Q.); (C.G.)
| | - Camille Gautier
- Platform Artificial Insemination and Embryo Transfer, Department for Small Animals and Horses, Vetmeduni Vienna, Veterinärplatz 1, 1210 Vienna, Austria; (C.T.C.O.); (M.K.); (J.P.Q.); (C.G.)
| | - Jörg Aurich
- Section for Obstetrics, Gynaecology and Andrology, Department for Small Animals and Horses, Vetmeduni Vienna, Veterinärplatz 1, 1210 Vienna, Austria;
| | - Christine Aurich
- Platform Artificial Insemination and Embryo Transfer, Department for Small Animals and Horses, Vetmeduni Vienna, Veterinärplatz 1, 1210 Vienna, Austria; (C.T.C.O.); (M.K.); (J.P.Q.); (C.G.)
- Correspondence:
| |
Collapse
|
15
|
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: 1.6] [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
|
16
|
Kisspeptin has an independent and direct effect on the pituitary gland in the mare. Theriogenology 2020; 157:199-209. [PMID: 32814247 DOI: 10.1016/j.theriogenology.2020.07.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 06/27/2020] [Accepted: 07/26/2020] [Indexed: 11/22/2022]
Abstract
To more clearly understand the equine gonadotrope response to kisspeptin and gonadotropin releasing hormone (GnRH), peripheral LH and FSH were quantified in diestrous mares after treatment with either equine kisspeptide (eKp-10, 0.5 mg iv), GnRH (25 μg iv), or a combination thereof every 4 h for 3 days. The following observations were made: 1) a diminished LH and FSH response to eKp-10 and GnRH was observed by Day 3, but was not different by treatment, 2) a decrease in basal LH concentration was observed from Day 1 to Day 3 for the eKp-10, but not the GnRH treated mares, 3) there was no change in basal FSH with either treatment. Additionally, pre-treatment with GnRH antagonist (antide 1.0 mg iv) eliminated any measurable change in LH after eKp-10 (1.0 mg iv) treatment. Both GnRH and kisspeptin are Gαq/11 coupled receptors, therefore quantifying the rise in intracellular calcium following treatment with cognate ligand allows simultaneous assessment of receptor activation. Direct stimulation of equine primary pituitary cells with GnRH and/or eKp-10 demonstrates three distinct populations of pituitary cells: one population responded to both eKp-10 and GnRH, a second, independent population, responded to only eKp-10, and a third population responded only to GnRH. These populations were confirmed using co-immunofluorescence of hemipituitaries from mares in diestrus. Although the rise in peripheral LH concentration elicited by eKp-10 is dependent on GnRH, this work suggests that kisspeptin also has a specific and direct effect on the equine gonadotrope, independent of GnRH.
Collapse
|
17
|
Merkley CM, Renwick AN, Shuping SL, Harlow K, Sommer JR, Nestor CC. Undernutrition reduces kisspeptin and neurokinin B expression in castrated male sheep. REPRODUCTION AND FERTILITY 2020; 1:1-13. [PMID: 35128420 PMCID: PMC8812452 DOI: 10.1530/raf-20-0025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 10/01/2020] [Indexed: 11/09/2022] Open
Abstract
Undernutrition impairs reproductive success through suppression of gonadotropin-releasing hormone (GnRH), and subsequently luteinizing hormone (LH), secretion. Given that kisspeptin and neurokinin B (NKB) neurons in the arcuate nucleus (ARC) of the hypothalamus are thought to play key stimulatory roles in the generation of GnRH/LH pulses, we hypothesized that feed restriction would reduce the ARC mRNA abundance and protein expression of kisspeptin and NKB in young, male sheep. Fourteen wethers (castrated male sheep five months of age) were either fed to maintain (FM; n = 6) pre-study body weight or feed-restricted (FR; n = 8) to lose 20% of pre-study body weight over 13 weeks. Throughout the study, weekly blood samples were collected and assessed for LH concentration using RIA. At Week 13 of the experiment, animals were killed, heads were perfused with 4% paraformaldehyde, and brain tissue containing the hypothalamus was collected, sectioned, and processed for detection of mRNA (RNAscope) and protein (immunohistochemistry) for kisspeptin and NKB. Mean LH was significantly lower and LH inter-pulse interval was significantly higher in FR wethers compared to FM wethers at the end of the experiment (Week 13). RNAscope analysis revealed significantly fewer cells expressing mRNA for kisspeptin and NKB in FR wethers compared to FM controls, and immunohistochemical analysis revealed significantly fewer immunopositive kisspeptin and NKB cells in FR wethers compared to FM wethers. Taken together, this data supports the idea that long-term feed restriction regulates GnRH/LH secretion through central suppression of kisspeptin and NKB in male sheep. LAY SUMMARY While undernutrition is known to impair reproduction at the level of the brain, the components responsible for this in the brain remain to be fully understood. Using male sheep we examined the effect of undernutrition on two stimulatory molecules in the brain critical for reproduction: kisspeptin and neurokinin B. Feed restriction for several weeks resulted in decreased luteinizing hormone in the blood indicating reproductive function was suppressed. In addition, undernutrition also reduced both kisspeptin and neurokinin B levels within a region of the brain involved in reproduction, the hypothalamus. Given that they have stimulatory roles in reproduction, we believe that undernutrition acts in the brain to reduce kisspeptin and neurokinin B levels leading to the reduction in luteinizing hormone secretion. In summary, long-term undernutrition inhibits reproductive function in sheep through suppression of kisspeptin and neurokinin B within the brain.
Collapse
Affiliation(s)
- Christina M Merkley
- Department of Animal Science, North Carolina State University, Raleigh, North Carolina, USA
| | - Allison N Renwick
- Department of Animal Science, North Carolina State University, Raleigh, North Carolina, USA
| | - Sydney L Shuping
- Department of Animal Science, North Carolina State University, Raleigh, North Carolina, USA
| | - KaLynn Harlow
- Department of Animal Science, North Carolina State University, Raleigh, North Carolina, USA
| | - Jeffrey R Sommer
- Department of Animal Science, North Carolina State University, Raleigh, North Carolina, USA
| | - Casey C Nestor
- Department of Animal Science, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
18
|
Zhang R, Nie H, Duan S, Yan P, Izaz A, Wang R, Zhou Y, Wu X. Cloning, characterisation and expression profile of kisspeptin1 and the kisspeptin1 receptor in the hypothalamic–pituitary–ovarian axis of Chinese alligator Alligator sinensis during the reproductive cycle. Reprod Fertil Dev 2020; 32:792-804. [DOI: 10.1071/rd19332] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 01/22/2020] [Indexed: 12/24/2022] Open
Abstract
Kisspeptin1 (Kiss1), a product of the Kiss1 gene, plays an important role in the regulation of reproduction in vertebrates by activating the Kiss1 receptor (Kiss1R) and its coexpression with gonadotrophin-releasing hormone (GnRH) in GnRH neurons. The purpose of this study was to clone the Kiss1 and Kiss1R genes found in the brain of Alligator sinensis and to explore their relationship with reproduction. The full-length cDNA of Kiss1 is 816bp, the open reading frame (ORF) is 417bp and the gene encodes a 138-amino acid precursor protein. The full-length cDNA of Kiss1R is 2348bp, the ORF is 1086bp and the gene encodes a 361-amino acid protein. Quantitative polymerase chain reaction showed that, except for Kiss1R expression in the hypothalamus, the expression of Kiss1 and Kiss1Rduring the reproductive period of A. sinensis was higher than that in the hypothalamus, pituitary gland and ovary during the hibernation period. The changes in GnRH2 mRNA in the hypothalamus were similar to those of GnRH1 and peaked during the reproductive period. This study confirms the existence of Kiss1 and Kiss1R in A. sinensis and the findings strongly suggest that Kiss1 and Kiss1R may participate in the regulation of GnRH secretion in the hypothalamus of alligators during the reproductive period. Furthermore, this is the first report of the full-length cDNA sequences of Kiss1 and Kiss1R in reptiles.
Collapse
|
19
|
Northup SL, Coffman EA, Strickland LG, Pohler KG, Daniel JA, Whitlock BK. Intravenous infusion of kisspeptin increased serum luteinizing hormone acutely and decreased serum follicle stimulating hormone chronically in prepubertal bull calves. Theriogenology 2019; 144:1-7. [PMID: 31881476 DOI: 10.1016/j.theriogenology.2019.12.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/15/2019] [Accepted: 12/19/2019] [Indexed: 12/01/2022]
Abstract
Kisspeptin (KP) is a hypothalamic neuropeptide that stimulates the secretion of gonadotropin releasing hormone. To determine the acute and chronic effects of KP on serum concentrations of luteinizing hormone (LH) and follicle stimulating hormone (FSH), prepubertal bull calves [12 ± 1 (SD) weeks of age; 96.5 ± 14.5 kg BW] were administered one of four treatments [0.0 (control; CON), 0.125 (L-KP), 0.25 (M-KP), or 0.5 (H-KP) μg of KP/kg BW/hour] by intravenous infusion for 76 h. Blood samples were collected every 15 min for the first (acute; 1-6 h; Day 1) and last (chronic; 71-76 h; Day 4) 6 h of the intravenous infusions. Serum concentrations of LH and FSH were determined by radioimmunoassay. For each day, effects of treatment, time, and interactions on LH and FSH concentrations and pulse parameters were analyzed using procedures for repeated measures with JMP Software (SAS Inst. Inc., Cary, NC). There was a treatment effect (P = 0.002) and a treatment × time interaction during Day 1 (P = 0.02) such that LH concentrations were greatest following administration of all doses of KP when compared to CON. However, there was no treatment effect (P = 0.57) or a treatment × time interaction during Day 4 (P = 0.20) on serum LH concentrations. There was a treatment by day interaction (P = 0.02) on mean serum FSH concentrations. Most notably, on Day 4 mean serum FSH concentrations during intravenous infusion of M-KP and H-KP doses were less than that of CON. There was a treatment by day interaction (P = 0.0054) on FSH pulse amplitude concentrations, such that intravenous infusion of all doses of KP on Day 4 decreased FSH pulse amplitudes. In conclusion, acute infusion of KP increased LH concentrations and chronic infusion of KP decreased FSH concentrations. Despite the potential suppression of the hypothalamic-pituitary-gonadal axis with chronic infusion of KP, there are likely applications of KP, KP analogs, or KP receptor agonists to hasten the onset of puberty in livestock.
Collapse
Affiliation(s)
- Samantha L Northup
- College of Veterinary Medicine, University of Tennessee, Knoxville, TN, USA; Department of Animal Science, University of Tennessee, Knoxville, TN, USA
| | - Elizabeth A Coffman
- School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Lew G Strickland
- College of Veterinary Medicine, University of Tennessee, Knoxville, TN, USA; Department of Animal Science, University of Tennessee, Knoxville, TN, USA
| | - Ky G Pohler
- Department of Animal Science, University of Tennessee, Knoxville, TN, USA
| | - Jay A Daniel
- School of Mathematical and Natural Sciences, Berry College, Mount Berry, GA, USA
| | - Brian K Whitlock
- College of Veterinary Medicine, University of Tennessee, Knoxville, TN, USA.
| |
Collapse
|
20
|
|
21
|
Maśko M, Domino M, Zdrojkowski Ł, Jasiński T, Matyba P, Zabielski R, Gajewski Z. Breeding management of mares in late reproductive age considering improvement of welfare. A review. JOURNAL OF ANIMAL AND FEED SCIENCES 2018. [DOI: 10.22358/jafs/100461/2018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
22
|
Scott CJ, Rose JL, Gunn AJ, McGrath BM. Kisspeptin and the regulation of the reproductive axis in domestic animals. J Endocrinol 2018; 240:JOE-18-0485.R1. [PMID: 30400056 DOI: 10.1530/joe-18-0485] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 10/09/2018] [Indexed: 11/08/2022]
Abstract
The control of reproductive processes involves the integration of a number of factors from the internal and external environment, with the final output signal of these processes being the pulsatile secretion of gonadotrophin releasing hormone (GnRH) from the hypothalamus. These factors include the feedback actions of sex steroids, feed intake and nutritional status, season/photoperiod, pheromones, age and stress. Understanding these factors and how they influence GnRH secretion and hence reproduction is important for the management of farm animals. There is evidence that the RF-amide neuropeptide, kisspeptin, may be involved in relaying the effects of these factors to the GnRH neurons. This paper will review the evidence from the common domestic animals (sheep, goats, cattle, horses and pigs), that kisspeptin neurons are i) regulated by the factors listed above, ii) contact GnRH neurons, and iii) involved in the regulation of GnRH/gonadotrophin secretion.
Collapse
Affiliation(s)
- Christopher J Scott
- C Scott, School of Biomedical Sciences, Charles Sturt University - Wagga Wagga Campus, Wagga Wagga, Australia
| | - Jessica L Rose
- J Rose, School of Biomedical Sciences, Charles Sturt University - Wagga Wagga Campus, Wagga Wagga, Australia
| | - Allan J Gunn
- A Gunn, School of Animal and Veterinary Sciences, Charles Sturt University - Wagga Wagga Campus, Wagga Wagga, Australia
| | - Briony M McGrath
- B McGrath, School of Biomedical Sciences, Charles Sturt University - Wagga Wagga Campus, Wagga Wagga, Australia
| |
Collapse
|
23
|
Effect of kisspeptin-10, LH and hCG on serum testosterone concentrations in stallions, donkeys and mules. Theriogenology 2017; 102:75-79. [DOI: 10.1016/j.theriogenology.2017.07.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 07/20/2017] [Accepted: 07/21/2017] [Indexed: 11/18/2022]
|
24
|
Beltramo M, Decourt C. Towards new strategies to manage livestock reproduction using kisspeptin analogs. Theriogenology 2017; 112:2-10. [PMID: 28916209 DOI: 10.1016/j.theriogenology.2017.08.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 08/29/2017] [Accepted: 08/30/2017] [Indexed: 12/16/2022]
Abstract
The discovery of the hypothalamic neuropeptide kisspeptin and its receptor (KISS1R) have dramatically improved our knowledge about the central mechanisms controlling reproduction. Kisspeptin neurons could be considered the hub where internal and external information controlling reproduction converge. The information is here elaborated and the command dispatched to GnRH neurons, the final output of the brain system controlling reproduction. Several studies have shown that in mammals administration of kisspeptin could finely modulate many aspects of reproduction from puberty to ovulation. For example in ewes kisspeptin infusion triggered ovulation during the non-breeding season and in prepubertal rat repeated injections advanced puberty onset. However, especially in livestock, the suboptimal pharmacological properties of endogenous kisspeptin, notably it short half-life and consequently its poor pharmacodynamics, fetters its use to experimental setting. To overcome this issue synthetic KISS1R agonists, mainly based on kisspeptin backbone, were created. Their more favorable pharmacological profile, longer half-life and duration of action, allowed to perform promising initial experiments for controlling ovulation and puberty. Additional experiments and further refinement of analogs would still be necessary to exploit fully the potential of targeting the kisspeptin system. Nevertheless, it is already clear that this new strategy may represent a breakthrough in the field of reproduction control.
Collapse
Affiliation(s)
- M Beltramo
- INRA, UMR85 Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, France; CNRS, UMR7247, F-37380 Nouzilly, France; Université François Rabelais de Tours, F-37041 Tours, France; IFCE, F-37380 Nouzilly, France.
| | - C Decourt
- INRA, UMR85 Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, France; CNRS, UMR7247, F-37380 Nouzilly, France; Université François Rabelais de Tours, F-37041 Tours, France; IFCE, F-37380 Nouzilly, France
| |
Collapse
|
25
|
McGrath BM, Scott CJ, Wynn PC, Loy J, Norman ST. Kisspeptin stimulates LH secretion but not ovulation in mares during vernal transition. Theriogenology 2016; 86:1566-1572. [DOI: 10.1016/j.theriogenology.2016.05.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 05/13/2016] [Accepted: 05/16/2016] [Indexed: 11/26/2022]
|
26
|
Tanco VM, Whitlock BK, Jones MA, Wilborn RR, Brandebourg TD, Foradori CD. Distribution and regulation of gonadotropin-releasing hormone, kisspeptin, RF-amide related peptide-3, and dynorphin in the bovine hypothalamus. PeerJ 2016; 4:e1833. [PMID: 27014517 PMCID: PMC4806599 DOI: 10.7717/peerj.1833] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 03/01/2016] [Indexed: 01/06/2023] Open
Abstract
Recent work has led to the hypothesis that kisspeptin/neurokinin B/dynorphin (KNDy) neurons in the arcuate nucleus (ARC) play a key role in gonadotropin-releasing hormone (GnRH) pulse generation and gonadal steroid feedback, with kisspeptin driving GnRH release and neurokinin B and dynorphin acting as pulse start and stop signals, respectively. A separate cell group, expressing RFamide-related peptide-3 (RFRP-3) has been shown to be a primary inhibitor of GnRH release. Very little is known regarding these cell groups in the bovine. In this study, we examined the relative immunoreactivity of kisspeptin, dynorphin, and RFRP-3 and their possible connectivity to GnRH neurons in the hypothalami of periestrus and diestrus bovine. While GnRH and RFRP-3 immunoreactivity were unchanged, kisspeptin and dynorphin immunoreactivity levels varied in relation to plasma progesterone concentrations and estrous status. Animals with higher plasma progesterone concentrations in diestrus had lower kisspeptin and increased dynorphin immunoreactivity in the ARC. The percentage of GnRH cells with kisspeptin or RFRP-3 fibers in close apposition did not differ between estrous stages. However, the proportions of GnRH cells with kisspeptin or RFRP-3 contacts (∼49.8% and ∼31.3%, respectively) suggest direct communication between kisspeptin and RFRP-3 cells to GnRH cells in the bovine. The data produced in this work support roles for kisspeptin and dynorphin, within the KNDy neural network, in controlling GnRH release over the ovarian cycle and conveying progesterone-negative feedback onto GnRH neurons in the bovine.
Collapse
Affiliation(s)
- Valeria M Tanco
- Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee-Knoxville , Knoxville, TN , United States
| | - Brian K Whitlock
- Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee-Knoxville , Knoxville, TN , United States
| | - Melaney A Jones
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University , Auburn, AL , United States
| | - Robyn R Wilborn
- Department of Clinical Sciences, College of Veterinary Medicine, Auburn University , Auburn, AL , United States
| | - Terry D Brandebourg
- Department of Animal Sciences, College of Agriculture, Auburn University , Auburn, AL , United States
| | - Chad D Foradori
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University , Auburn, AL , United States
| |
Collapse
|
27
|
Hua W, Luo L, Tian Y, Song M, Liu Y, Cui P, Song S, Jiang S, Li F, Fang F. Analysis of the serum concentrations of kisspeptin and neurokinin B in the geese during reproductive cycle and their localisation in the ovary. Anim Reprod Sci 2014; 151:78-84. [DOI: 10.1016/j.anireprosci.2014.09.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Revised: 09/02/2014] [Accepted: 09/12/2014] [Indexed: 11/27/2022]
|
28
|
Decourt C, Caraty A, Briant C, Guillaume D, Lomet D, Chesneau D, Lardic L, Duchamp G, Reigner F, Monget P, Dufourny L, Beltramo M, Dardente H. Acute Injection and Chronic Perfusion of Kisspeptin Elicit Gonadotropins Release but Fail to Trigger Ovulation in the Mare1. Biol Reprod 2014; 90:36. [DOI: 10.1095/biolreprod.113.114157] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
29
|
Zhou S, Holmes MM, Forger NG, Goldman BD, Lovern MB, Caraty A, Kalló I, Faulkes CG, Coen CW. Socially regulated reproductive development: Analysis of GnRH-1 and kisspeptin neuronal systems in cooperatively breeding naked mole-rats (Heterocephalus glaber). J Comp Neurol 2013; 521:3003-29. [DOI: 10.1002/cne.23327] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 02/20/2013] [Accepted: 02/26/2013] [Indexed: 12/18/2022]
Affiliation(s)
- Shuzhi Zhou
- Reproductive Neurobiology; Division of Women's Health; School of Medicine; King's College London; London; UK
| | | | | | | | | | - Alain Caraty
- INRA; Physiology of Reproduction; Nouzilly; France
| | | | | | - Clive W. Coen
- Reproductive Neurobiology; Division of Women's Health; School of Medicine; King's College London; London; UK
| |
Collapse
|
30
|
Mechaly AS, Viñas J, Piferrer F. The kisspeptin system genes in teleost fish, their structure and regulation, with particular attention to the situation in Pleuronectiformes. Gen Comp Endocrinol 2013; 188:258-68. [PMID: 23624122 DOI: 10.1016/j.ygcen.2013.04.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 04/16/2013] [Indexed: 10/26/2022]
Abstract
It is well established that Kisspeptin regulates the onset of puberty in vertebrates through stimulation of the secretion of gonadotropin-releasing hormones. However, the function of kisspeptin in peripheral tissues and in other functions is still poorly understood. Recently, the evolution and distribution of kisspeptin genes in vertebrates has been clarified. In contrast to placental mammals, which have a single gene for the ligand (Kiss) and for the receptor (Kissr), fish may have up to three Kiss genes and up to four Kissr genes because of genome duplications. However, information on the genomic structure of the piscine kiss and kissr genes is still scarce. Furthermore, when data from several species is taken together, interspecific differences in the expression of kiss and kissr during the reproductive cycle are found. Here, we discuss data gathered from several fish species, but mainly from two flatfishes, the Senegalese sole and the Atlantic halibut, to address general questions on kiss gene structure, regulation and function. Flatfish are among the most derived fish species and the two species referred to above have only one ligand and one receptor, probably because of the genome reduction observed in Pleuronectiformes. However, gene analysis shows that both species have an alternative splicing mechanism based on intron retention, but the functions of the alternative isoforms are unclear. In the Senegalese sole, sex-related differences in the temporal and spatial expression of kiss and kissr were observed during a whole reproductive cycle. In addition, recent studies suggested that kisspeptin system gene expression is correlated to energy balance and reproduction. This suggests that kisspeptin signaling may involve different sources of information to synchronize important biological functions in vertebrates, including reproduction. We propose a set of criteria to facilitate the comparison of kiss and kissr gene expression data across species.
Collapse
Affiliation(s)
- Alejandro S Mechaly
- Institut de Ciències del Mar, Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain.
| | | | | |
Collapse
|
31
|
Okamura H, Yamamura T, Wakabayashi Y. Kisspeptin as a master player in the central control of reproduction in mammals: an overview of kisspeptin research in domestic animals. Anim Sci J 2013; 84:369-381. [PMID: 23607315 DOI: 10.1111/asj.12056] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 01/16/2013] [Indexed: 11/30/2022]
Abstract
The hypothalamo-pituitary-gonadal (HPG) axis is the regulatory system for reproduction in mammals. Because secretion of gonadotropin-releasing hormone (GnRH) into the portal vessels is the final step at which the brain controls gonadal activities, the GnRH neuronal system had been thought to be central to the HPG axis. A newly discovered neural peptide, kisspeptin, has opened a new era in reproductive neuroendocrinology. As shown in a variety of mammals, kisspeptin is a potent endogenous secretagogue of GnRH, and the kisspeptin neuronal system governs both the pulsatile GnRH secretion that drives folliculogenesis, spermatogenesis and steroidogenesis, and the GnRH surge that triggers ovulation in females. The kisspeptin neuronal system is therefore considered a master player in the central control of mammalian reproduction, and kisspeptin and related substances could therefore be valuable for the development of novel strategies for the management of fertility in farm animals. To this end, the present review aimed to summarize the current research on kisspeptin signaling with a focus on domestic animals such as sheep, goats, cattle, pigs and horses.
Collapse
Affiliation(s)
- Hiroaki Okamura
- Animal Physiology Research Unit, National Institute of Agrobiological Sciences, Tsukuba, Ibaraki, Japan
| | - Takashi Yamamura
- Animal Physiology Research Unit, National Institute of Agrobiological Sciences, Tsukuba, Ibaraki, Japan
| | - Yoshihiro Wakabayashi
- Animal Physiology Research Unit, National Institute of Agrobiological Sciences, Tsukuba, Ibaraki, Japan
| |
Collapse
|
32
|
Lehman MN, Hileman SM, Goodman RL. Neuroanatomy of the kisspeptin signaling system in mammals: comparative and developmental aspects. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 784:27-62. [PMID: 23550001 DOI: 10.1007/978-1-4614-6199-9_3] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Our understanding of kisspeptin and its actions depends, in part, on a detailed knowledge of the neuroanatomy of the kisspeptin signaling system in the brain. In this chapter, we will review our current knowledge of the distribution of kisspeptin cells, fibers, and receptors in the mammalian brain, including the development, phenotype, and projections of different kisspeptin subpopulations. A fairly consistent picture emerges from this analysis. There are two major groups of kisspeptin cell bodies: a large number in the arcuate nucleus (ARC) and a smaller collection in the rostral periventricular area of the third ventricle (RP3V) of rodents and preoptic area (POA) of non-rodents. Both sets of neurons project to GnRH cell bodies, which contain Kiss1r, and the ARC kisspeptin population also projects to GnRH axons in the median eminence. ARC kisspeptin neurons contain neurokinin B and dynorphin, while a variable percentage of those cells in the RP3V of rodents contain galanin and/or dopamine. Neurokinin B and dynorphin have been postulated to contribute to the control of GnRH pulses and sex steroid negative feedback, while the role of galanin and dopamine in rostral kisspeptin neurons is not entirely clear. Kisspeptin neurons, fibers, and Kiss1r are found in other areas, including widespread areas outside the hypothalamus, but their physiological role(s) in these regions remains to be determined.
Collapse
Affiliation(s)
- Michael N Lehman
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS, USA.
| | | | | |
Collapse
|
33
|
The effects of kisspeptin on gonadotropin release in non-human mammals. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 784:63-87. [PMID: 23550002 DOI: 10.1007/978-1-4614-6199-9_4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The Kiss1 gene encodes a 145-amino acid pre-peptide, kisspeptin, which is cleaved into smaller peptides of 54, 14, 13, and 10 amino acids. This chapter reviews in detail the effects of kisspeptin on gonadotropin secretion in non-human mammals. Studies of kisspeptin's effects have included both acute and chronic administration regimens via a number of administration routes. Acute kisspeptin stimulates gonadotropin secretion in a wide range of species of non-human mammals, including rats, mice, hamsters, sheep, pigs, goats, cows, horses, and monkeys. In general, the stimulatory effect of kisspeptin treatment is more pronounced for LH than FSH secretion. Kisspeptin is thought to exert its stimulatory effects on LH and FSH release via stimulation of GnRH release from the hypothalamus, since pre--administration of a GnRH antagonist prevents kisspeptin's stimulation of gonadotropin secretion. Although the kisspeptin receptor is also expressed on anterior pituitary cells of some species, and incubation of anterior pituitary cells with high concentrations of kisspeptin can stimulate in vitro LH release, the contribution of direct effects of kisspeptin on the pituitary is thought to be negligible in vivo. Continuous kisspeptin administration results in reduced sensitivity to the effects of kisspeptin, in some species. This desensitization is thought to occur at the level of the kisspeptin receptor, since the response of the pituitary gland to exogenous GnRH is maintained. Overall, the findings discussed in this chapter are invaluable to the understanding of the reproductive role of kisspeptin and the potential therapeutic uses of kisspeptin for the treatment of fertility disorders.
Collapse
|
34
|
Magee C, Bruemmer J, Nett T, Squires E, Clay C. Kisspeptide in the estrous mare: Is it an appropriate ovulation-inducing agent? Theriogenology 2012; 78:1987-96. [DOI: 10.1016/j.theriogenology.2012.07.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Revised: 07/11/2012] [Accepted: 07/12/2012] [Indexed: 01/23/2023]
|
35
|
Kaur KK, Allahbadia G, Singh M. Kisspeptins in human reproduction-future therapeutic potential. J Assist Reprod Genet 2012; 29:999-1011. [PMID: 23015158 PMCID: PMC3492584 DOI: 10.1007/s10815-012-9856-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Accepted: 08/27/2012] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVE Kisspeptins (Kps), were first found to regulate the hypothalamopituitary-gonadal axis (HPG) axis in 2003, when two groups-demonstrated that mutations of GPR54 causes idiopathic hypogonadotropic hypogonadism (IHH) characterized by delayed puberty. Objective of this review is to highlight both animal and human discoveries in KISS1/GPR54 system in last decade and extrapolate the therapeutic potential in humans from till date human studies. DESIGN A systematic review of international scientific literature by a search of PUBMED and the authors files was done for Kp in reproduction, metabolic control & signal transduction. SETTING None Patient(s): In human studies--normal subjects patients with HH, or HA. MAIN OUTCOME MEASURES Effects of Kp on puberty, brain sexual maturation, regulation of GnRH secretion, metabolic control of GnRH Neurons (N). RESULTS Kps/GPR54 are critical for brain sexual maturation, puberty and regulation of reproduction. Kps have been implicated in mediating signals to GnRH N--positive and negative feedback, metabolic input. Ability of Kp neurons to coordinate signals impinging on the HPG axis makes it one of most important regulators of reproductive axis since GnRH N's lack many receptors, with Kp neurons serving as upstream modulators. CONCLUSIONS Kps have proven as pivotal regulators of the reproduction, with the ability to integrate signals from both internal and external sources. Knowledge about signaling mechanisms involved in Kp stimulation of GnRH and with human studies has made it possible that therapeutically available Kp agonists/antagonists may be used for treatment of delayed puberty/HH, Hypothalamic amenorrhea and in prevention of spread of malignant ovarian/gonadal malignancies along with uses in some eating disorders.
Collapse
Affiliation(s)
- Kulvinder Kochar Kaur
- Dr Kulvinder Kaur Centre for Human Reproduction, 721, G.T.B. Nagar, Jalandhar, 144001 Punjab India
| | - Gautam Allahbadia
- Rotunda-A Centre for Human reproduction, 672, Kalpak Garden, Perry Cross Road, Near Otter’s Club, Bandra (W), 400040 Mumbai, India
| | - Mandeep Singh
- Swami Satyanand Hospital, Near Nawi Kachehri, Baradri, Ladowali Road, Jalandhar, Punjab India
| |
Collapse
|
36
|
Caraty A, Decourt C, Briant C, Beltramo M. Kisspeptins and the reproductive axis: potential applications to manage reproduction in farm animals. Domest Anim Endocrinol 2012; 43:95-102. [PMID: 22533939 DOI: 10.1016/j.domaniend.2012.03.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Revised: 03/13/2012] [Accepted: 03/14/2012] [Indexed: 10/28/2022]
Abstract
Kisspeptins (Kp) are a family of neuropeptides produced mainly by two hypothalamic neuronal cell populations. They have recently emerged as a major regulator of the gonadotropin axis and their action is located upstream of the gonadotropin-releasing hormone (GnRH) cell population. In less than 10 yr a growing body of literature has demonstrated the involvement of these peptides in most, if not all, aspects of reproductive axis maturation and function. In contrast to these abundant basic research studies, few experiments have evaluated the potential application of Kp as tools to manipulate reproduction in domestic animals. In mammals, exogenous Kp administration potently stimulates gonadotropin secretion. This action is exerted mainly, if not exclusively, through the stimulation of GnRH release. Intravenous, intraperitoneal, or subcutaneous administration of Kp induced a robust and rapid increase in plasma gonadotropins (luteinizing hormone [LH] and follicle-stimulating hormone [FSH]). However, this stimulatory effect is of short duration. Prolonged LH and FSH release over several hours can be achieved only when Kp are given as repeated multiple bolus or as an infusion. Kp administration was used in two experimental models, ewe and pony mare, with the aim of inducing well-timed and synchronized ovulations. During the breeding season, progesterone-synchronized ewes were given an intravenous infusion of Kp starting 30 h after the removal of progesterone implants. An LH surge was induced in all Kp-treated animals within 2 h of infusion onset. In contrast, in pony mares a constant infusion of Kp for 3 d in the the late follicular phase was unable to induce synchronized ovulation. Another set of studies showed that Kp could be used to activate reproductive function in acyclic animals. Pulsatile administration of Kp in prepubertal ewe lambs was shown to activate ovarian function, leading to enhanced ovarian steroidogenesis, stimulation of LH preovulatory surge, and ovulation. In anestrous ewes, an intravenous infusion of a low dose of Kp induced an immediate and sustained release of gonadotropins, followed a few hours later by an LH surge. This hormonal pattern mimicked hormonal changes normally observed during the estrous cycle follicular phase and was associated with a high percentage of ovulating animals (80%). In summary, exogenous administration of Kp appears to be a new tool to manipulate reproduction. However, optimal doses and periods of treatment should be defined for each species, and the development of powerful analogs or long-term release formulations is necessary before large-scale applications in domestic animals could be envisaged.
Collapse
Affiliation(s)
- A Caraty
- Unité Mixte de Recherche 6175, Physiologie de la Reproduction et des Comportements (INRA/CNRS/Université de Tours/Haras Nationaux), Nouzilly, France.
| | | | | | | |
Collapse
|
37
|
Pinilla L, Aguilar E, Dieguez C, Millar RP, Tena-Sempere M. Kisspeptins and Reproduction: Physiological Roles and Regulatory Mechanisms. Physiol Rev 2012; 92:1235-316. [DOI: 10.1152/physrev.00037.2010] [Citation(s) in RCA: 529] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Procreation is essential for survival of species. Not surprisingly, complex neuronal networks have evolved to mediate the diverse internal and external environmental inputs that regulate reproduction in vertebrates. Ultimately, these regulatory factors impinge, directly or indirectly, on a final common pathway, the neurons producing the gonadotropin-releasing hormone (GnRH), which stimulates pituitary gonadotropin secretion and thereby gonadal function. Compelling evidence, accumulated in the last few years, has revealed that kisspeptins, a family of neuropeptides encoded by the Kiss1 gene and produced mainly by neuronal clusters at discrete hypothalamic nuclei, are pivotal upstream regulators of GnRH neurons. As such, kisspeptins have emerged as important gatekeepers of key aspects of reproductive maturation and function, from sexual differentiation of the brain and puberty onset to adult regulation of gonadotropin secretion and the metabolic control of fertility. This review aims to provide a comprehensive account of the state-of-the-art in the field of kisspeptin physiology by covering in-depth the consensus knowledge on the major molecular features, biological effects, and mechanisms of action of kisspeptins in mammals and, to a lesser extent, in nonmammalian vertebrates. This review will also address unsolved and contentious issues to set the scene for future research challenges in the area. By doing so, we aim to endow the reader with a critical and updated view of the physiological roles and potential translational relevance of kisspeptins in the integral control of reproductive function.
Collapse
Affiliation(s)
- Leonor Pinilla
- Department of Cell Biology, Physiology and Immunology, University of Córdoba; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III; and Instituto Maimónides de Investigaciones Biomédicas, Córdoba, Spain; Department of Physiology, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain; and Centre for Integrative Physiology, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Enrique Aguilar
- Department of Cell Biology, Physiology and Immunology, University of Córdoba; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III; and Instituto Maimónides de Investigaciones Biomédicas, Córdoba, Spain; Department of Physiology, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain; and Centre for Integrative Physiology, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Carlos Dieguez
- Department of Cell Biology, Physiology and Immunology, University of Córdoba; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III; and Instituto Maimónides de Investigaciones Biomédicas, Córdoba, Spain; Department of Physiology, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain; and Centre for Integrative Physiology, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Robert P. Millar
- Department of Cell Biology, Physiology and Immunology, University of Córdoba; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III; and Instituto Maimónides de Investigaciones Biomédicas, Córdoba, Spain; Department of Physiology, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain; and Centre for Integrative Physiology, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Manuel Tena-Sempere
- Department of Cell Biology, Physiology and Immunology, University of Córdoba; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III; and Instituto Maimónides de Investigaciones Biomédicas, Córdoba, Spain; Department of Physiology, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain; and Centre for Integrative Physiology, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
38
|
Caraty A, Blomenröhr M, Vogel GMT, Lomet D, Briant C, Beltramo M. RF9 powerfully stimulates gonadotrophin secretion in the ewe: evidence for a seasonal threshold of sensitivity. J Neuroendocrinol 2012; 24:725-36. [PMID: 22283564 DOI: 10.1111/j.1365-2826.2012.02283.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
GPR147 and its endogenous ligands, RFRPs, are emerging as important actors in hypothalamic-pituitary axis control. The role of this system would be to inhibit gonadotrophin secretion. However, data on the subject are contradictory. The discovery of RF9 (adamantanecarbonyl-RF-2-NH(2)), a GPR147 antagonist, prompted us to use this new tool to further investigate this system in the ewe. Accordingly, we tested the effect of i.c.v. administration of RF9 on gonadotrophin secretion in the ewe during anoestrous and the breeding season. Intracerebroventricular injections of RF9 (from 50-450 nmol) caused a clear elevation in peripheral blood plasma luteinising hormone (LH) concentrations. The effect of RF9 on LH was more pronounced during the anoestrous season. Furthermore, peripheral administration of RF9 as a bolus (2.1, 6.2 and 12.4 μmol per ewe) or as a constant i.v. infusion (2.1, 6.2, 12.4 and 18.6 μmol/h per ewe) to anoestrous acyclic ewes induced a sustained increase in LH plasma concentrations. A pharmacokinetic study showed that RF9 (12.4 μmol bolus i.v.) has an effective half life of 5.5 h in the plasma. Conversely, RF9 is not detectable in the cerebrospinal fluid, suggesting that it does not cross the blood-brain barrier. The increase in LH plasma concentrations induced by RF9 was blocked by previous administration of 1.3 μmol per ewe of gondotrophin-releasing hormone (GnRH) antagonist Teverelix. This suggests that GnRH is involved in the stimulatory effect of RF9 on gonadotrophin secretion. Finally, no variation in LH plasma concentrations could be detected in ovariectomised ewes injected either i.c.v. or i.v. with RFRP3 (VPNLPQRF-NH(2)). The lack of effect of RFRP3 in our experimental setting suggests that the mechanisms involved in RF9 action are probably more complex than previously assumed. Our results indicate that delivery of RF9 in the ewe greatly increases gondadotrophin secretion in both the oestrus and anoestrus season, suggesting a potential new way of controlling reproduction in mammals.
Collapse
Affiliation(s)
- A Caraty
- UMR 7247, Physiologie de la Reproduction et des Comportements (INRA/CNRS/Université Tours/Haras Nationaux), 37380 Nouzilly, France.
| | | | | | | | | | | |
Collapse
|
39
|
Abstract
Seasonal mammals typically of temperate or boreal habitats use the predictable annual cycle of daylength to initiate a suite of physiological and behavioural changes in anticipation of adverse environmental winter conditions, unfavourable for survival and reproduction. Daylength is encoded as the duration of production of the pineal hormone melatonin, but how the melatonin signal is decoded has been elusive. From the studies carried out in birds and mammals together with the advent of technologies such as microarray analysis of gene expression, progress has been achieved to demystify how seasonal physiology is regulated in response to the duration of melatonin signalling. The critical tissue for the action of melatonin is the pars tuberalis (PT) where melatonin receptors are located. At the molecular level, regulation of cyclic adenosine monophosphate (cAMP) signalling in this tissue is likely to be a key event for melatonin action, either an acute inhibitory action or sensitization of this pathway by prolonged stimulation of melatonin receptors reflecting durational melatonin presence. Melatonin action at the PT has been shown to have both positive and negative effects on gene transcription, incorporating components of the circadian clock as part of the mechanism of decoding the melatonin signal and regulating thyrotrophin-stimulating hormone (TSH) expression, a key output hormone of the PT. Microarray analysis of gene expression of PT tissue exposed to long and short photoperiods has identified important new genes that may be regulated by melatonin and contributing to the seasonal regulation of TSH production by this tissue. In the brain, tanycytes lining the third ventricle of the hypothalamus and regulation of thyroid hormone synthesis by PT-derived TSH in these cells are now established as an important component of the pathway leading to seasonal changes in physiology. Beyond the tanycyte, identified changes in gene expression for neuropeptides, receptors and other signalling molecules pinpoint some of the areas of the brain, the hypothalamus in particular, that are likely to be involved in the regulation of seasonal physiology.
Collapse
Affiliation(s)
- Perry Barrett
- Rowett Institute for Nutrition and Health, University of Aberdeen, Aberdeen, UK.
| | | |
Collapse
|
40
|
Redmond JS, Macedo GG, Velez IC, Caraty A, Williams GL, Amstalden M. Kisspeptin activates the hypothalamic–adenohypophyseal–gonadal axis in prepubertal ewe lambs. Reproduction 2011; 141:541-8. [DOI: 10.1530/rep-10-0467] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The onset of puberty in mammals involves an increase in the pulsatile release of GNRH and LH. The KISS1 gene is essential for pubertal development, and its product, kisspeptin, stimulates the release of LH. The objective of this study was to determine the effects of kisspeptin in the hypothalamic–adenohypophyseal–gonadal axis of prepubertal ewe lambs. Ewe lambs (28 weeks of age) were treated intravenously with saline (control, n=6) or kisspeptin (20 μg kisspeptin; n=6) every hour for 24 h. Kisspeptin stimulated pulse-like release of LH within 15 min following injections, and increased the frequency and amplitude of LH pulses, and mean circulating concentrations of LH and estradiol. A surge-like release of LH was observed in four kisspeptin-treated lambs beginning 17 h after the onset of treatment, and all four lambs had elevated circulating concentrations of progesterone within 5 days post-treatment. However, circulating concentrations of progesterone decreased within 2 days after the initial rise in three of the four ewe lambs, indicating that induced luteal activity was of short duration. The proportion of lambs that were pubertal (defined by circulating concentrations of progesterone above 1 ng/ml for at least 7 days) by 35 weeks of age (8/11) and the mean age at puberty (32±1 weeks) for those reaching puberty within the experimental period did not differ between treatments. Results support a role for kisspeptin in the activation of the hypothalamic–adenohypophyseal axis leading to the onset of puberty in ewe lambs.
Collapse
|
41
|
Hrabovszky E, Molnár CS, Sipos MT, Vida B, Ciofi P, Borsay BA, Sarkadi L, Herczeg L, Bloom SR, Ghatei MA, Dhillo WS, Kalló I, Liposits Z. Sexual dimorphism of kisspeptin and neurokinin B immunoreactive neurons in the infundibular nucleus of aged men and women. Front Endocrinol (Lausanne) 2011; 2:80. [PMID: 22654828 PMCID: PMC3356022 DOI: 10.3389/fendo.2011.00080] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Accepted: 11/08/2011] [Indexed: 12/02/2022] Open
Abstract
The secretory output of gonadotropin-releasing hormone (GnRH) neurons is critically influenced by peptidergic neurons synthesizing kisspeptins (KP) and neurokinin B (NKB) in the hypothalamic infundibular nucleus (Inf). These cells mediate negative feedback effects of sex steroids on the reproductive axis. While negative feedback is lost in postmenopausal women, it is partly preserved by the sustained testosterone secretion in aged men. We hypothesized that the different reproductive physiology of aged men and women is reflected in morphological differences of KP and NKB neurons. This sexual dimorphism was studied with immunohistochemistry in hypothalamic sections of aged human male (≥50 years) and female (>55 years) subjects. KP and NKB cell bodies of the Inf were larger in females. The number of KP cell bodies, the density of KP fibers, and the incidence of their contacts on GnRH neurons were much higher in aged women compared with men. The number of NKB cell bodies was only slightly higher in women and there was no sexual dimorphism in the regional density of NKB fibers and the incidence of their appositions onto GnRH cells. The incidences of NKB cell bodies, fibers, and appositions onto GnRH neurons exceeded several-fold those of KP-IR elements in men. More NKB than KP inputs to GnRH cells were also present in women. Immunofluorescent studies identified only partial overlap between KP and NKB axons. KP and NKB were colocalized in higher percentages of afferents to GnRH neurons in women compared with men. Most of these sex differences might be explained with the lack of estrogen negative feedback in aged women, whereas testosterone can continue to suppress KP, and to a lesser extent, NKB synthesis in men. Overall, sex differences in reproductive physiology of aged humans were reflected in the dramatic sexual dimorphism of the KP system, with significantly higher incidences of KP-IR neurons, fibers and inputs to GnRH neurons in aged females vs. males.
Collapse
Affiliation(s)
- Erik Hrabovszky
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of SciencesBudapest, Hungary
- *Correspondence: Erik Hrabovszky, Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, 43 Szigony Street, Budapest 1083, Hungary. e-mail:
| | - Csilla S. Molnár
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of SciencesBudapest, Hungary
| | - Máté T. Sipos
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of SciencesBudapest, Hungary
| | - Barbara Vida
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of SciencesBudapest, Hungary
| | | | - Beáta A. Borsay
- Department of Forensic Medicine, University of DebrecenDebrecen, Hungary
| | - László Sarkadi
- Department of Forensic Medicine, University of DebrecenDebrecen, Hungary
| | - László Herczeg
- Department of Forensic Medicine, University of DebrecenDebrecen, Hungary
| | - Stephen R. Bloom
- Department of Investigative Medicine, Hammersmith Hospital, Imperial College LondonLondon, UK
| | - Mohammad A. Ghatei
- Department of Investigative Medicine, Hammersmith Hospital, Imperial College LondonLondon, UK
| | - Waljit S. Dhillo
- Department of Investigative Medicine, Hammersmith Hospital, Imperial College LondonLondon, UK
| | - Imre Kalló
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of SciencesBudapest, Hungary
- Department of Neuroscience, Pázmány Péter Catholic UniversityBudapest, Hungary
| | - Zsolt Liposits
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of SciencesBudapest, Hungary
- Department of Neuroscience, Pázmány Péter Catholic UniversityBudapest, Hungary
| |
Collapse
|
42
|
Kirby HR, Maguire JJ, Colledge WH, Davenport AP. International Union of Basic and Clinical Pharmacology. LXXVII. Kisspeptin receptor nomenclature, distribution, and function. Pharmacol Rev 2010; 62:565-78. [PMID: 21079036 PMCID: PMC2993257 DOI: 10.1124/pr.110.002774] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Kisspeptins are members of the Arg-Phe amide family of peptides, which have been identified as endogenous ligands for a G-protein-coupled receptor encoded by a gene originally called GPR54 (also known as AXOR12 or hOT7T175). After this pairing, the gene has been renamed KISS1R. The International Union of Basic and Clinical Pharmacology Committee on Receptor Nomenclature and Drug Classification recommends that the official name for the receptor is the kisspeptin receptor to follow the convention of naming the receptor protein after the endogenous ligand. The endogenous ligand was initially called metastin, after its role as a metastasis suppressor, and is now referred to as kisspeptin-54 (KP-54), a C-terminally amidated 54-amino acid peptide cleaved from the 145-amino acid gene product. Shorter C-terminal cleavage fragments [KP-14, KP-13 and KP-10 (the smallest active fragment)] are also biologically active. Both receptor and peptide are widely expressed in human, rat, and mouse; the receptor sequence shares more than 80% homology in these species. Activation of the kisspeptin receptor by kisspeptin is via coupling to G(q/11) and the phospholipase C pathway, causing Ca(2+) mobilization. Mutations in the KISS1R gene result in hypogonadotropic hypogonadotropism, and targeted disruption of Kiss1r in mice reproduces this phenotype, which led to the discovery of the remarkable ability of the kisspeptin receptor to act as a molecular switch for puberty. In addition to regulating the reproductive axis, the kisspeptin receptor is also implicated in cancer, placentation, diabetes, and the cardiovascular system.
Collapse
Affiliation(s)
- Helen R Kirby
- Clinical Pharmacology Unit, University of Cambridge, Addenbrookes Hospital, Cambridge, United Kingdom
| | | | | | | |
Collapse
|
43
|
Desroziers E, Mikkelsen J, Simonneaux V, Keller M, Tillet Y, Caraty A, Franceschini I. Mapping of kisspeptin fibres in the brain of the pro-oestrous rat. J Neuroendocrinol 2010; 22:1101-12. [PMID: 20673302 DOI: 10.1111/j.1365-2826.2010.02053.x] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Kisspeptins are a family of small peptides that play a key role in the neuroendocrine regulation of the reproductive function through neural pathways that have not yet been completely identified. The present study aimed to investigate the distribution of kisspeptin neurone fibres in the female rat brain by comparing precisely the immunoreactive pattern obtained with two antibodies: one specifically directed against kisspeptin-52 (Kp-52), the longest isoform, and the other directed against kisspeptin-10 (Kp-10), whose sequence is common to all putative mature isoforms. With both antibodies, immunoreactive cell bodies were exclusively observed in the arcuate nucleus, and immunoreactive fibres were confined to the septo-preoptico-hypothalamic continuum of the brain. Fibres were observed in the preoptic area, the diagonal band of Broca, the septohypothalamic area, the anteroventral periventricular, suprachiasmatic, supraoptic, paraventricular and periventricular nuclei, the dorsal border of the ventromedian nucleus, the dorsomedial and arcuate nuclei, and the median eminence. In the latter structure, varicose fibres were mainly distributed in the internal layer and were detected to a lesser extent throughout the external layer, including around the deeper part of the infundibular recess. Most regions of immunoreactive cells and fibres matched perfectly for the two antibodies. However, fibres in the dorsolateral septum, anterior fornix, accumbens nucleus and the lateral bed nucleus of the stria terminalis were only recognised by antibody anti-Kp-10, suggesting that anti-Kp-10 may recognise a wider range of kisspeptin isoforms than anti-Kp-52 or cross-react with molecules other than kisspeptin in rat tissue. Overall, these results illustrate the variety of projection sites of kisspeptin neurones in the rat and suggest that these peptides play a role in different functions.
Collapse
Affiliation(s)
- E Desroziers
- UMR 6175 INRA/CNRS/Université de Tours/Haras Nationaux, Nouzilly, France
| | | | | | | | | | | | | |
Collapse
|
44
|
Anatomy of the kisspeptin neural network in mammals. Brain Res 2010; 1364:90-102. [PMID: 20858464 DOI: 10.1016/j.brainres.2010.09.020] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2010] [Revised: 09/02/2010] [Accepted: 09/02/2010] [Indexed: 01/17/2023]
Abstract
Kisspeptin has been recognized as a key regulator of GnRH secretion during puberty and adulthood, conveying the feedback influence of endogenous gonadal steroids onto the GnRH system. Understanding the functional roles of this peptide depends on knowledge of the anatomical framework in which it acts, including the location of kisspeptin-expressing cells in the brain and their connections. In this paper, we review current data on the anatomy of the kisspeptin neuronal network, including its colocalization with gonadal steroid hormone receptors, anatomical sites of interaction with the GnRH system, and recent evidence of neurochemical heterogeneity among different kisspeptin neuronal populations. Evidence to date suggests that kisspeptin cells in mammals comprise an interconnected network, with reciprocal connections both within and between separate cell populations, and with GnRH neurons. At the same time, there is more functional and anatomical heterogeneity in this system than originally thought, and many unanswered questions remain concerning anatomical relationships of kisspeptin neurons with other neuroendocrine and neural systems in the brain.
Collapse
|
45
|
Abstract
Kisspeptin (a product of the Kiss1 gene) and its receptor (GPR54 or Kiss1r) have emerged as key players in the regulation of reproduction. Mutations in humans or genetically targeted deletions in mice of either Kiss1 or Kiss1r cause profound hypogonadotropic hypogonadism. Neurons that express Kiss1/kisspeptin are found in discrete nuclei in the hypothalamus, as well as other brain regions in many vertebrates, and their distribution, regulation, and function varies widely across species. Kisspeptin neurons directly innervate and stimulate GnRH neurons, which are the final common pathway through which the brain regulates reproduction. Kisspeptin neurons are sexually differentiated with respect to cell number and transcriptional activity in certain brain nuclei, and some kisspeptin neurons express other cotransmitters, including dynorphin and neurokinin B (whose physiological significance is unknown). Kisspeptin neurons express the estrogen receptor and the androgen receptor, and these cells are direct targets for the action of gonadal steroids in both male and female animals. Kisspeptin signaling in the brain has been implicated in mediating the negative feedback action of sex steroids on gonadotropin secretion, generating the preovulatory GnRH/LH surge, triggering and guiding the tempo of sexual maturation at puberty, controlling seasonal reproduction, and restraining reproductive activity during lactation. Kisspeptin signaling may also serve diverse functions outside of the classical realm of reproductive neuroendocrinology, including the regulation of metastasis in certain cancers, vascular dynamics, placental physiology, and perhaps even higher-order brain function.
Collapse
Affiliation(s)
- Amy E Oakley
- Department of Physiology and Biophysics, University of Washington, Box 356460, 1705 NE Pacific Street, Health Sciences Building Room BB604, Seattle, Washington 98195-6460, USA.
| | | | | |
Collapse
|