1
|
Dardente H, Lomet D, Robert V, Lasserre O, Gonzalez AA, Mialhe X, Beltramo M. Photoperiod, but not progesterone, has a strong impact upon the transcriptome of the medio-basal hypothalamus in female goats and ewes. Mol Cell Endocrinol 2024; 588:112216. [PMID: 38556161 DOI: 10.1016/j.mce.2024.112216] [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: 11/23/2023] [Revised: 03/11/2024] [Accepted: 03/24/2024] [Indexed: 04/02/2024]
Abstract
Photoperiod is the main environmental driver of seasonal responses in organisms living at temperate and polar latitudes. Other external cues such as food and temperature, and internal cues including hormones, intervene to fine-tune phasing of physiological functions to the solar year. In mammals, the medio-basal hypothalamus (MBH) is the key integrator of these cues, which orchestrates a wide array of seasonal functions, including breeding. Here, using RNAseq and RT-qPCR, we demonstrate that molecular components of the photoperiodic response previously identified in ewes are broadly conserved in does (female goats, Capra hircus), with a common core of ∼50 genes. This core group can be defined as the "MBH seasonal trancriptome", which includes key players of the pars tuberalis-tanycytes neuroendocrine retrograde pathway that governs intra-MBH photoperiodic switches of triiodothyronine (T3) production (Tshb, Eya3, Dio2 and SlcO1c1), the two histone methyltransferases Suv39H2 and Ezh2 and the secreted protein Vmo1. Prior data in ewes revealed that T3 and estradiol (E2), both key hormones for the proper timing of seasonal breeding, differentially impact the MBH seasonal transcriptome, and identified cellular and molecular targets through which these hormones might act. In contrast, information regarding the potential impact of progesterone (P4) upon the MBH transcriptome was nonexistent. Here, we demonstrate that P4 has no discernible transcriptional impact in either does or ewes. Taken together, our data show that does and ewes possess a common core set of photoperiod-responsive genes in the MBH and conclusively demonstrate that P4 is not a key regulator of the MBH transcriptome.
Collapse
Affiliation(s)
- Hugues Dardente
- INRAE, CNRS, Université de Tours, PRC, 37380, Nouzilly, France.
| | - Didier Lomet
- INRAE, CNRS, Université de Tours, PRC, 37380, Nouzilly, France
| | - Vincent Robert
- INRAE, CNRS, Université de Tours, PRC, 37380, Nouzilly, France
| | | | - Anne-Alicia Gonzalez
- MGX-Montpellier GenomiX, Univ. Montpellier, CNRS, INSERM, 34094, Montpellier, France
| | - Xavier Mialhe
- MGX-Montpellier GenomiX, Univ. Montpellier, CNRS, INSERM, 34094, Montpellier, France
| | | |
Collapse
|
2
|
Uehara SK, Nishiike Y, Maeda K, Karigo T, Kuraku S, Okubo K, Kanda S. Identification of the FSH-RH as the other gonadotropin-releasing hormone. Nat Commun 2024; 15:5342. [PMID: 38937445 PMCID: PMC11211334 DOI: 10.1038/s41467-024-49564-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 06/10/2024] [Indexed: 06/29/2024] Open
Abstract
In vertebrates, folliculogenesis and ovulation are regulated by two distinct pituitary gonadotropins: follicle-stimulating hormone (FSH) and luteinizing hormone (LH). Currently, there is an intriguing consensus that a single hypothalamic neurohormone, gonadotropin-releasing hormone (GnRH), regulates the secretion of both FSH and LH, although the required timing and functions of FSH and LH are different. However, recent studies in many non-mammalian vertebrates indicated that GnRH is dispensable for FSH function. Here, by using medaka as a model teleost, we successfully identify cholecystokinin as the other gonadotropin regulator, FSH-releasing hormone (FSH-RH). Our histological and in vitro analyses demonstrate that hypothalamic cholecystokinin-expressing neurons directly affect FSH cells through the cholecystokinin receptor, Cck2rb, thereby increasing the expression and release of FSH. Remarkably, the knockout of this pathway minimizes FSH expression and results in a failure of folliculogenesis. Here, we propose the existence of the "dual GnRH model" in vertebrates that utilize both FSH-RH and LH-RH.
Collapse
Affiliation(s)
- Shun Kenny Uehara
- Atmosphere and Ocean Research Institute, The University of Tokyo, Chiba, Japan
| | - Yuji Nishiike
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Kazuki Maeda
- Atmosphere and Ocean Research Institute, The University of Tokyo, Chiba, Japan
| | - Tomomi Karigo
- Kennedy Krieger Institute, Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shigehiro Kuraku
- Laboratory for Phyloinformatics, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
- Molecular Life History Laboratory, Department of Genomics and Evolutionary Biology, National Institute of Genetics, Mishima, Japan
| | - Kataaki Okubo
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Shinji Kanda
- Atmosphere and Ocean Research Institute, The University of Tokyo, Chiba, Japan.
| |
Collapse
|
3
|
Męczekalski B, Niwczyk O, Battipaglia C, Troia L, Kostrzak A, Bala G, Maciejewska-Jeske M, Genazzani AD, Luisi S. Neuroendocrine disturbances in women with functional hypothalamic amenorrhea: an update and future directions. Endocrine 2024; 84:769-785. [PMID: 38062345 PMCID: PMC11208264 DOI: 10.1007/s12020-023-03619-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 11/17/2023] [Indexed: 01/31/2024]
Abstract
Functional hypothalamic amenorrhea (FHA) is one of the most common causes of both primary and secondary amenorrhea in women of reproductive age. It is characterized by chronic anovulation and the absence of menses that appear as a result of stressors such as eating disorders, excessive exercise, or psychological distress. FHA is presumed to be a functional disruption in the pulsatile secretion of hypothalamic gonadotropin-releasing hormone, which in turn impairs the release of gonadotropin. Hypoestrogenism is observed due to the absence of ovarian follicle recruitment. Numerous neurotransmitters have been identified which play an important role in the regulation of the hypothalamic-pituitary-ovarian axis and of which the impairment would contribute to developing FHA. In this review we summarize the most recent advances in the identification of contributing neuroendocrine disturbances and relevant contributors to the development of FHA.
Collapse
Affiliation(s)
- Błażej Męczekalski
- Department of Gynecological Endocrinology, Poznan University of Medical Sciences, Poznan, Poland.
| | - Olga Niwczyk
- Department of Gynecological Endocrinology, Poznan University of Medical Sciences, Poznan, Poland
| | - Christian Battipaglia
- Gynecological Endocrinology Center, Department of Obstetrics and Gynecology, University of Modena and Reggio Emilia, Modena, Italy
| | - Libera Troia
- Department of Gynecology and Obstetrics, Maggiore della Carità Hospital, University of Eastern Piedmont, Novara, Italy
| | - Anna Kostrzak
- Department of Gynecological Endocrinology, Poznan University of Medical Sciences, Poznan, Poland
| | - Gregory Bala
- UCD School of Medicine University College Dublin, D04 V1W8, Dublin, Ireland
| | | | - Alessandro D Genazzani
- Gynecological Endocrinology Center, Department of Obstetrics and Gynecology, University of Modena and Reggio Emilia, Modena, Italy
| | - Stefano Luisi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| |
Collapse
|
4
|
Nakamura S, Sasaki T, Uenoyama Y, Inoue N, Nakanishi M, Yamada K, Morishima A, Suzumura R, Kitagawa Y, Morita Y, Ohkura S, Tsukamura H. Raphe glucose-sensing serotonergic neurons stimulate KNDy neurons to enhance LH pulses via 5HT2CR: rat and goat studies. Sci Rep 2024; 14:10190. [PMID: 38702366 PMCID: PMC11068885 DOI: 10.1038/s41598-024-58470-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 03/29/2024] [Indexed: 05/06/2024] Open
Abstract
Dysfunction of central serotonergic neurons is known to cause depressive disorders in humans, who often show reproductive and/or glucose metabolism disorders. This study examined whether dorsal raphe (DR) serotonergic neurons sense high glucose availability to upregulate reproductive function via activating hypothalamic arcuate (ARC) kisspeptin neurons (= KNDy neurons), a dominant stimulator of gonadotropin-releasing hormone (GnRH)/gonadotropin pulses, using female rats and goats. RNA-seq and histological analysis revealed that stimulatory serotonin-2C receptor (5HT2CR) was mainly expressed in the KNDy neurons in female rats. The serotonergic reuptake inhibitor administration into the mediobasal hypothalamus (MBH), including the ARC, significantly blocked glucoprivic suppression of luteinizing hormone (LH) pulses and hyperglycemia induced by intravenous 2-deoxy-D-glucose (2DG) administration in female rats. A local infusion of glucose into the DR significantly increased in vivo serotonin release in the MBH and partly restored LH pulses and hyperglycemia in the 2DG-treated female rats. Furthermore, central administration of serotonin or a 5HT2CR agonist immediately evoked GnRH pulse generator activity, and central 5HT2CR antagonism blocked the serotonin-induced facilitation of GnRH pulse generator activity in ovariectomized goats. These results suggest that DR serotonergic neurons sense high glucose availability to reduce gluconeogenesis and upregulate reproductive function by activating GnRH/LH pulse generator activity in mammals.
Collapse
Affiliation(s)
- Sho Nakamura
- Laboratory of Animal Production Science, Graduate School of Bioagricultural Sciences, Nagoya University, Togo-cho, Aichi, 470-0151, Japan
| | - Takuya Sasaki
- Laboratory of Animal Production Science, Graduate School of Bioagricultural Sciences, Nagoya University, Togo-cho, Aichi, 470-0151, Japan
| | - Yoshihisa Uenoyama
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, 464-8601, Japan
| | - Naoko Inoue
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, 464-8601, Japan
| | - Marina Nakanishi
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, 464-8601, Japan
| | - Koki Yamada
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, 464-8601, Japan
| | - Ai Morishima
- Laboratory of Animal Production Science, Graduate School of Bioagricultural Sciences, Nagoya University, Togo-cho, Aichi, 470-0151, Japan
| | - Reika Suzumura
- Laboratory of Animal Production Science, Graduate School of Bioagricultural Sciences, Nagoya University, Togo-cho, Aichi, 470-0151, Japan
| | - Yuri Kitagawa
- Laboratory of Animal Production Science, Graduate School of Bioagricultural Sciences, Nagoya University, Togo-cho, Aichi, 470-0151, Japan
| | - Yasuhiro Morita
- Laboratory of Animal Production Science, Graduate School of Bioagricultural Sciences, Nagoya University, Togo-cho, Aichi, 470-0151, Japan
| | - Satoshi Ohkura
- Laboratory of Animal Production Science, Graduate School of Bioagricultural Sciences, Nagoya University, Togo-cho, Aichi, 470-0151, Japan
| | - Hiroko Tsukamura
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, 464-8601, Japan.
| |
Collapse
|
5
|
Athar F, Karmani M, Templeman N. Metabolic hormones are integral regulators of female reproductive health and function. Biosci Rep 2024; 44:BSR20231916. [PMID: 38131197 PMCID: PMC10830447 DOI: 10.1042/bsr20231916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 11/29/2023] [Accepted: 12/21/2023] [Indexed: 12/23/2023] Open
Abstract
The female reproductive system is strongly influenced by nutrition and energy balance. It is well known that food restriction or energy depletion can induce suppression of reproductive processes, while overnutrition is associated with reproductive dysfunction. However, the intricate mechanisms through which nutritional inputs and metabolic health are integrated into the coordination of reproduction are still being defined. In this review, we describe evidence for essential contributions by hormones that are responsive to food intake or fuel stores. Key metabolic hormones-including insulin, the incretins (glucose-dependent insulinotropic polypeptide and glucagon-like peptide-1), growth hormone, ghrelin, leptin, and adiponectin-signal throughout the hypothalamic-pituitary-gonadal axis to support or suppress reproduction. We synthesize current knowledge on how these multifaceted hormones interact with the brain, pituitary, and ovaries to regulate functioning of the female reproductive system, incorporating in vitro and in vivo data from animal models and humans. Metabolic hormones are involved in orchestrating reproductive processes in healthy states, but some also play a significant role in the pathophysiology or treatment strategies of female reproductive disorders. Further understanding of the complex interrelationships between metabolic health and female reproductive function has important implications for improving women's health overall.
Collapse
Affiliation(s)
- Faria Athar
- Department of Biology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Muskan Karmani
- Department of Biology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Nicole M. Templeman
- Department of Biology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| |
Collapse
|
6
|
Sang L, Sun S, Wang J, Gao C, Chen D, Xie X. Dual effects of gonadotropin-inhibitory hormone on testicular development in prepubertal Minxinan Black rabbits ( Oryctolagus cuniculus). Front Vet Sci 2024; 11:1320452. [PMID: 38328257 PMCID: PMC10847550 DOI: 10.3389/fvets.2024.1320452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 01/11/2024] [Indexed: 02/09/2024] Open
Abstract
Gonadotropin-inhibitory hormone (GnIH) is a neurohormone that not only suppresses reproduction at the brain level but also regulates steroidogenesis and gametogenesis at the gonad level. However, its function in gonadal physiology has received little attention in rabbits. The main objective of this study was to evaluate the effects of GnIH on testicular development and function in prepubertal Minxinan Black rabbits (Oryctolagus cuniculus). In the present study, we investigated the serum reproductive hormone concentration, testicular parameters, morphology of seminiferous tubules, apoptosis of testicular cells, and expression of reproductive-related genes in male prepubertal Minxinan Black rabbits intraperitoneally administered with 0, 0.5, 5, or 50 μg quail GnIH-related peptides (qGnIH) for 10 days. Compared with the vehicle, administration with 5 μg of qGnIH downregulated the serum testosterone concentration and mRNA levels of spermatogenic genes (PCNA, FSHR, INHβA, HSF1, and AR) and upregulated the apoptosis rate of testicular cells; administration with 50 μg of qGnIH decreased the serum testosterone concentration and hypothalamic GnIH gene mRNA level and increased the serum LH concentration, pituitary LHβ gene mRNA level, testicular weight, gonadosomatic index (GSI), and spermatogenic cell layer thickness. It is concluded that GnIH could exert dual actions on testicular development depending on the male prepubertal rabbits receiving different intraperitoneal doses.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiping Xie
- Fujian Key Laboratory of Animal Genetics and Breeding, Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, China
| |
Collapse
|
7
|
Bigambo FM, Wang D, Sun J, Ding X, Li X, Gao B, Wu D, Gu W, Zhang M, Wang X. Association between Urinary BPA Substitutes and Precocious Puberty among Girls: A Single-Exposure and Mixed Exposure Approach from a Chinese Case-Control Study. TOXICS 2023; 11:905. [PMID: 37999557 PMCID: PMC10675366 DOI: 10.3390/toxics11110905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 10/27/2023] [Accepted: 11/03/2023] [Indexed: 11/25/2023]
Abstract
There is an argument that BPA substitutes may have the same or more deleterious health effects as BPA due to their structural similarity. This study explored the association between urinary BPA substitutes and precocious puberty among girls by including 120 girls with precocious puberty (cases) aged 2-10 years enrolled at Nanjing Children's Hospital Department of Endocrinology in China between April 2021 to September 2021 and 145 healthy girls (controls) recruited from a primary school. Logistic regression was used to evaluate the effect of single exposures, and Bayesian kernel machine regression (BKMR) and quantile-based g-computation were used for the mixed effect. In the multivariate logistic regression, BPS (bisphenol S), TBBPA (tetrabromobisphenol A), and BPFL (bisphenol-FL) were significantly associated with increased risk of precocious puberty (odds ratio (OR) = 1.75, 95% confidence interval (CI): 1.13, 2.76, p = 0.014), (OR = 1.46, CI: 1.06, 2.05; p = 0.023), and (OR = 1.47, CI: 1.01, 2.18; p = 0.047), respectively. The BMKR and quantile-based g-computation models revealed consistent associations for single exposures and there was insufficient evidence for the associations of the mixed exposure of bisphenols with precocious puberty. In conclusion, BPA substitutes such as BPS, TBBPA, and BPFL may be associated with an increased risk of precocious puberty in girls.
Collapse
Affiliation(s)
- Francis Manyori Bigambo
- Department of Endocrinology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China; (F.M.B.); (D.W.); (W.G.)
| | - Dandan Wang
- Department of Endocrinology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China; (F.M.B.); (D.W.); (W.G.)
| | - Jian Sun
- Department of Emergency, Pediatric Intensive Care Unit, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China;
| | - Xinliang Ding
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China; (X.D.); (X.L.); (B.G.); (D.W.)
- Wuxi Center for Disease Control and Prevention, The Affiliated Wuxi Center for Disease Control and Prevention, Nanjing Medical University, Wuxi 214023, China
- Research Base for Environment and Health in Wuxi, Chinese Center for Disease Control and Prevention, Wuxi 214023, China
| | - Xiuzhu Li
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China; (X.D.); (X.L.); (B.G.); (D.W.)
- Wuxi Center for Disease Control and Prevention, The Affiliated Wuxi Center for Disease Control and Prevention, Nanjing Medical University, Wuxi 214023, China
- Research Base for Environment and Health in Wuxi, Chinese Center for Disease Control and Prevention, Wuxi 214023, China
| | - Beibei Gao
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China; (X.D.); (X.L.); (B.G.); (D.W.)
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Di Wu
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China; (X.D.); (X.L.); (B.G.); (D.W.)
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Wei Gu
- Department of Endocrinology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China; (F.M.B.); (D.W.); (W.G.)
| | - Mingzhi Zhang
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China; (X.D.); (X.L.); (B.G.); (D.W.)
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Xu Wang
- Department of Endocrinology, Children’s Hospital of Nanjing Medical University, Nanjing 210008, China; (F.M.B.); (D.W.); (W.G.)
| |
Collapse
|
8
|
Uenoyama Y, Tsukamura H. KNDy neurones and GnRH/LH pulse generation: Current understanding and future aspects. J Neuroendocrinol 2023; 35:e13285. [PMID: 37232103 DOI: 10.1111/jne.13285] [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: 01/04/2023] [Revised: 03/31/2023] [Accepted: 04/26/2023] [Indexed: 05/27/2023]
Abstract
Uncovering the central mechanism underlying mammalian reproduction is warranted to develop new therapeutic approaches for reproductive disorders in humans and domestic animals. The present study focused on the role of arcuate kisspeptin neurones (also known as KNDy neurones) as an intrinsic gonadotropin-releasing hormone (GnRH) pulse generator, which plays a fundamental role in mammalian reproduction via the stimulation of pituitary gonadotropin synthesis and release and thereby in gametogenesis and steroidogenesis in the gonads of mammals. We also discuss the mechanism that inhibits pulsatile GnRH/gonadotropin release under a negative energy balance, considering that reproductive disorders often occur during malnutrition in humans and livestock.
Collapse
Affiliation(s)
| | - Hiroko Tsukamura
- Laboratory of Animal Reproduction, Nagoya University, Nagoya, Japan
| |
Collapse
|
9
|
Uenoyama Y, Inoue N, Tsukamura H. Kisspeptin and lactational anestrus: Current understanding and future prospects. Peptides 2023; 166:171026. [PMID: 37230188 DOI: 10.1016/j.peptides.2023.171026] [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: 03/03/2023] [Revised: 05/18/2023] [Accepted: 05/22/2023] [Indexed: 05/27/2023]
Abstract
Lactational anestrus, characterized by the suppression of pulsatile gonadotropin-releasing hormone (GnRH)/luteinizing hormone (LH) release, would be a strategic adaptation to ensure survival by avoiding pregnancy during lactation in mammals. In the present article, we first provide a current understanding of the central regulation of reproduction in mammals, i.e., a fundamental role of arcuate kisspeptin neurons in mammalian reproduction by driving GnRH/LH pulses. Second, we discuss the central mechanism inhibiting arcuate Kiss1 (encoding kisspeptin) expression and GnRH/LH pulses during lactation with a focus on suckling stimulus, negative energy balance due to milk production, and the role of circulating estrogen in rats. We also discuss upper regulators that control arcuate kisspeptin neurons in rats during the early and late lactation periods based on the findings obtained by a lactating rat model. Finally, we discuss potential reproductive technology for the improvement of reproductive performance in milking cows.
Collapse
Affiliation(s)
- Yoshihisa Uenoyama
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan.
| | - Naoko Inoue
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Hiroko Tsukamura
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| |
Collapse
|
10
|
Li Y, Zhang H, Wang Y, Li D, Chen H. Advances in circadian clock regulation of reproduction. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 137:83-133. [PMID: 37709382 DOI: 10.1016/bs.apcsb.2023.02.008] [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: 03/05/2023]
Abstract
The mammalian circadian clock is an endogenously regulated oscillator that is synchronized with solar time and cycle within a 24-h period. The circadian clock exists not only in the suprachiasmatic nucleus (SCN) of the hypothalamus, a central pacemaker of the circadian clock system, but also in numerous peripheral tissues known as peripheral circadian oscillators. The SCN and peripheral circadian oscillators mutually orchestrate the diurnal rhythms of various physiological and behavioral processes in a hierarchical manner. In the past two decades, peripheral circadian oscillators have been identified and their function has been determined in the mammalian reproductive system and its related endocrine glands, including the hypothalamus, pituitary gland, ovaries, testes, uterus, mammary glands, and prostate gland. Increasing evidence indicates that both the SCN and peripheral circadian oscillators play discrete roles in coordinating reproductive processes and optimizing fertility in mammals. The present study reviews recent evidence on circadian clock regulation of reproductive function in the hypothalamic-pituitary-gonadal axis and reproductive system. Additionally, we elucidate the effects of chronodisruption (as a result of, for example, shift work, jet lag, disrupted eating patterns, and sleep disorders) on mammalian reproductive performance from multiple aspects. Finally, we propose potential behavioral changes or pharmaceutical strategies for the prevention and treatment of reproductive disorders from the perspective of chronomedicine. Conclusively, this review will outline recent evidence on circadian clock regulation of reproduction, providing novel perspectives on the role of the circadian clock in maintaining normal reproductive functions and in diseases that negatively affect fertility.
Collapse
Affiliation(s)
- Yating Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, P.R. China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling, Shaanxi, P.R. China
| | - Haisen Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, P.R. China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling, Shaanxi, P.R. China
| | - Yiqun Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, P.R. China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling, Shaanxi, P.R. China
| | - Dan Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, P.R. China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling, Shaanxi, P.R. China
| | - Huatao Chen
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, P.R. China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling, Shaanxi, P.R. China.
| |
Collapse
|
11
|
Uenoyama Y, Tsuchida H, Nagae M, Inoue N, Tsukamura H. Opioidergic pathways and kisspeptin in the regulation of female reproduction in mammals. Front Neurosci 2022; 16:958377. [PMID: 36033602 PMCID: PMC9404872 DOI: 10.3389/fnins.2022.958377] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
Endogenous opioid peptides have attracted attention as critical neuropeptides in the central mechanism regulating female reproduction ever since the discovery that arcuate dynorphin neurons that coexpress kisspeptin and neurokinin B (NKB), which are also known as kisspeptin/neurokinin B/dynorphin (KNDy) neurons, play a role as a master regulator of pulsatile gonadotropin-releasing hormone (GnRH) release in mammals. In this study, we first focus on the role of dynorphin released by KNDy neurons in the GnRH pulse generation. Second, we provide a historical overview of studies on endogenous opioid peptides. Third, we discuss how endogenous opioid peptides modulate tonic GnRH/gonadotropin release in female mammals as a mediator of inhibitory internal and external cues, such as ovarian steroids, nutritional status, or stress, on reproduction. Then, we discuss the role of endogenous opioid peptides in GnRH surge generation in female mammals.
Collapse
|
12
|
López-Ojeda W, Hurley RA. Kisspeptin in the Limbic System: New Insights Into Its Neuromodulatory Roles. J Neuropsychiatry Clin Neurosci 2022; 34:190-195. [PMID: 35921618 DOI: 10.1176/appi.neuropsych.20220087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Wilfredo López-Ojeda
- Veterans Affairs Mid-Atlantic Mental Illness Research, Education, and Clinical Center and Research and Academic Affairs Service Line, W.G. Hefner Veterans Affairs Medical Center, Salisbury, N.C. (López-Ojeda, Hurley); Departments of Psychiatry and Behavioral Medicine (López-Ojeda, Hurley) and Radiology (Hurley), Wake Forest School of Medicine, Winston-Salem, N.C.; Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston (Hurley)
| | - Robin A Hurley
- Veterans Affairs Mid-Atlantic Mental Illness Research, Education, and Clinical Center and Research and Academic Affairs Service Line, W.G. Hefner Veterans Affairs Medical Center, Salisbury, N.C. (López-Ojeda, Hurley); Departments of Psychiatry and Behavioral Medicine (López-Ojeda, Hurley) and Radiology (Hurley), Wake Forest School of Medicine, Winston-Salem, N.C.; Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston (Hurley)
| |
Collapse
|
13
|
Stincic TL, Kelly MJ. Estrogenic regulation of reproduction and energy homeostasis by a triumvirate of hypothalamic arcuate neurons. J Neuroendocrinol 2022; 34:e13145. [PMID: 35581942 DOI: 10.1111/jne.13145] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 03/31/2022] [Accepted: 04/15/2022] [Indexed: 11/29/2022]
Abstract
Pregnancy is energetically demanding and therefore, by necessity, reproduction and energy balance are inextricably linked. With insufficient or excessive energy stores a female is liable to suffer complications during pregnancy or produce unhealthy offspring. Gonadotropin-releasing hormone neurons are responsible for initiating both the pulsatile and subsequent surge release of luteinizing hormone to control ovulation. Meticulous work has identified two hypothalamic populations of kisspeptin (Kiss1) neurons that are critical for this pattern of release. The involvement of the hypothalamus is unsurprising because its quintessential function is to couple the endocrine and nervous systems, coordinating energy balance and reproduction. Estrogens, more specifically 17β-estradiol (E2 ), orchestrate the activity of a triumvirate of hypothalamic neurons within the arcuate nucleus (ARH) that govern the physiological underpinnings of these behavioral dynamics. Arising from a common progenitor pool, these cells differentiate into ARH kisspeptin, pro-opiomelanocortin (POMC), and agouti related peptide/neuropeptide Y (AgRP) neurons. Although the excitability of all these subpopulations is subject to genomic and rapid estrogenic regulation, Kiss1 neurons are the most sensitive, reflecting their integral function in female fertility. Based on the premise that E2 coordinates autonomic functions around reproduction, we review recent findings on how Kiss1 neurons interact with gonadotropin-releasing hormone, AgRP and POMC neurons, as well as how the rapid membrane-initiated and intracellular signaling cascades activated by E2 in these neurons are critical for control of homeostatic functions supporting reproduction. In particular, we highlight how Kiss1 and POMC neurons conspire to inhibit AgRP neurons and diminish food motivation in service of reproductive success.
Collapse
Affiliation(s)
- Todd L Stincic
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, USA
| | - Martin J Kelly
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, USA
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| |
Collapse
|
14
|
Hu KL, Chen Z, Li X, Cai E, Yang H, Chen Y, Wang C, Ju L, Deng W, Mu L. Advances in clinical applications of kisspeptin-GnRH pathway in female reproduction. Reprod Biol Endocrinol 2022; 20:81. [PMID: 35606759 PMCID: PMC9125910 DOI: 10.1186/s12958-022-00953-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 04/30/2022] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Kisspeptin is the leading upstream regulator of pulsatile and surge Gonadotrophin-Releasing Hormone secretion (GnRH) in the hypothalamus, which acts as the key governor of the hypothalamic-pituitary-ovary axis. MAIN TEXT Exogenous kisspeptin or its receptor agonist can stimulate GnRH release and subsequent physiological gonadotropin secretion in humans. Based on the role of kisspeptin in the hypothalamus, a broad application of kisspeptin and its receptor agonist has been recently uncovered in humans, including central control of ovulation, oocyte maturation (particularly in women at a high risk of ovarian hyperstimulation syndrome), test for GnRH neuronal function, and gatekeepers of puberty onset. In addition, the kisspeptin analogs, such as TAK-448, showed promising agonistic activity in healthy women as well as in women with hypothalamic amenorrhoea or polycystic ovary syndrome. CONCLUSION More clinical trials should focus on the therapeutic effect of kisspeptin, its receptor agonist and antagonist in women with reproductive disorders, such as hypothalamic amenorrhoea, polycystic ovary syndrome, and endometriosis.
Collapse
Affiliation(s)
- Kai-Lun Hu
- Center for Reproductive Medicine, Peking University Third Hospital, No.49 Huayuan North Road, Haidian District, Beijing, People's Republic of China, 100191
- Zhejiang MedicalTech Therapeutics Company, No.665 Yumeng Road, Wenzhou, People's Republic of China, 325200
| | - Zimiao Chen
- Department of Endocrinology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China, 325000
| | - Xiaoxue Li
- Zhejiang MedicalTech Therapeutics Company, No.665 Yumeng Road, Wenzhou, People's Republic of China, 325200
| | - Enci Cai
- Department of Nutrition and Food Science, College of Food, Agricultural and Natural Resource Sciences, University of Minnesota, Twin Cities, Minneapolis, MN, 55455, USA
| | - Haiyan Yang
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China, 325000
| | - Yi Chen
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China, 325000
| | - Congying Wang
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China, 325000
| | - Liping Ju
- Zhejiang MedicalTech Therapeutics Company, No.665 Yumeng Road, Wenzhou, People's Republic of China, 325200
| | - Wenhai Deng
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China, 325006.
| | - Liangshan Mu
- Zhejiang MedicalTech Therapeutics Company, No.665 Yumeng Road, Wenzhou, People's Republic of China, 325200.
| |
Collapse
|
15
|
Moore AM, Coolen LM, Lehman MN. In vivo imaging of the GnRH pulse generator reveals a temporal order of neuronal activation and synchronization during each pulse. Proc Natl Acad Sci U S A 2022; 119:e2117767119. [PMID: 35110409 PMCID: PMC8833213 DOI: 10.1073/pnas.2117767119] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/30/2021] [Indexed: 01/08/2023] Open
Abstract
A hypothalamic pulse generator located in the arcuate nucleus controls episodic release of gonadotropin-releasing hormone (GnRH) and luteinizing hormone (LH) and is essential for reproduction. Recent evidence suggests this generator is composed of arcuate "KNDy" cells, the abbreviation based on coexpression of kisspeptin, neurokinin B, and dynorphin. However, direct visual evidence of KNDy neuron activity at a single-cell level during a pulse is lacking. Here, we use in vivo calcium imaging in freely moving female mice to show that individual KNDy neurons are synchronously activated in an episodic manner, and these synchronized episodes always precede LH pulses. Furthermore, synchronization among KNDy cells occurs in a temporal order, with some subsets of KNDy cells serving as "leaders" and others as "followers" during each synchronized episode. These results reveal an unsuspected temporal organization of activation and synchronization within the GnRH pulse generator, suggesting that different subsets of KNDy neurons are activated at pulse onset than afterward during maintenance and eventual termination of each pulse. Further studies to distinguish KNDy "leader" from "follower" cells is likely to have important clinical significance, since regulation of pulsatile GnRH secretion is essential for normal reproduction and disrupted in pathological conditions such as polycystic ovary syndrome and hypothalamic amenorrhea.
Collapse
Affiliation(s)
- Aleisha M Moore
- Brain Health Research Institute, Kent State University, Kent, OH 44242;
- Department of Biological Sciences, Kent State University, Kent, OH 44242
| | - Lique M Coolen
- Brain Health Research Institute, Kent State University, Kent, OH 44242
- Department of Biological Sciences, Kent State University, Kent, OH 44242
| | - Michael N Lehman
- Brain Health Research Institute, Kent State University, Kent, OH 44242
- Department of Biological Sciences, Kent State University, Kent, OH 44242
| |
Collapse
|
16
|
Rønnekleiv OK, Qiu J, Kelly MJ. Hypothalamic Kisspeptin Neurons and the Control of Homeostasis. Endocrinology 2022; 163:bqab253. [PMID: 34953135 PMCID: PMC8758343 DOI: 10.1210/endocr/bqab253] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Indexed: 12/27/2022]
Abstract
Hypothalamic kisspeptin (Kiss1) neurons provide indispensable excitatory transmission to gonadotropin-releasing hormone (GnRH) neurons for the coordinated release of gonadotropins, estrous cyclicity, and ovulation. But maintaining reproductive functions is metabolically demanding so there must be a coordination with multiple homeostatic functions, and it is apparent that Kiss1 neurons play that role. There are 2 distinct populations of hypothalamic Kiss1 neurons, namely arcuate nucleus (Kiss1ARH) neurons and anteroventral periventricular and periventricular nucleus (Kiss1AVPV/PeN) neurons in rodents, both of which excite GnRH neurons via kisspeptin release but are differentially regulated by ovarian steroids. Estradiol (E2) increases the expression of kisspeptin in Kiss1AVPV/PeN neurons but decreases its expression in Kiss1ARH neurons. Also, Kiss1ARH neurons coexpress glutamate and Kiss1AVPV/PeN neurons coexpress gamma aminobutyric acid (GABA), both of which are upregulated by E2 in females. Also, Kiss1ARH neurons express critical metabolic hormone receptors, and these neurons are excited by insulin and leptin during the fed state. Moreover, Kiss1ARH neurons project to and excite the anorexigenic proopiomelanocortin neurons but inhibit the orexigenic neuropeptide Y/Agouti-related peptide neurons, highlighting their role in regulating feeding behavior. Kiss1ARH and Kiss1AVPV/PeN neurons also project to the preautonomic paraventricular nucleus (satiety) neurons and the dorsomedial nucleus (energy expenditure) neurons to differentially regulate their function via glutamate and GABA release, respectively. Therefore, this review will address not only how Kiss1 neurons govern GnRH release, but how they control other homeostatic functions through their peptidergic, glutamatergic and GABAergic synaptic connections, providing further evidence that Kiss1 neurons are the key neurons coordinating energy states with reproduction.
Collapse
Affiliation(s)
- Oline K Rønnekleiv
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR 97239, USA
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Jian Qiu
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR 97239, USA
| | - Martin J Kelly
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR 97239, USA
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| |
Collapse
|
17
|
Regulation of stress response on the hypothalamic-pituitary-gonadal axis via gonadotropin-inhibitory hormone. Front Neuroendocrinol 2022; 64:100953. [PMID: 34757094 DOI: 10.1016/j.yfrne.2021.100953] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/16/2021] [Accepted: 10/24/2021] [Indexed: 11/21/2022]
Abstract
Under stressful condition, reproductive function is impaired due to the activation of various components of the hypothalamic-pituitaryadrenal (HPA) axis, which can suppress the activity of the hypothalamic-pituitary-gonadal (HPG) axis at multiple levels. A hypothalamic neuropeptide, gonadotropin-inhibitory hormone (GnIH) is a key negative regulator of reproduction that governs the HPG axis. Converging lines of evidence have suggested that different stress types and their duration, such as physical or psychological, and acute or chronic, can modulate the GnIH system. To clarify the sensitivity and reactivity of the GnIH system in response to stress, we summarize and critically review the available studies that investigated the effects of various stressors, such as restraint, nutritional/metabolic and social stress, on GnIH expression and/or its neuronal activity leading to altered HPG action. In this review, we focus on GnIH as the potential novel mediator responsible for stress-induced reproductive dysfunction.
Collapse
|
18
|
Deletion of Stim1 in Hypothalamic Arcuate Nucleus Kiss1 Neurons Potentiates Synchronous GCaMP Activity and Protects against Diet-Induced Obesity. J Neurosci 2021; 41:9688-9701. [PMID: 34654752 DOI: 10.1523/jneurosci.0622-21.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 06/29/2021] [Accepted: 10/07/2021] [Indexed: 12/17/2022] Open
Abstract
Kisspeptin (Kiss1) neurons are essential for reproduction, but their role in the control of energy balance and other homeostatic functions remains unclear. High-frequency firing of hypothalamic arcuate Kiss1 (Kiss1ARH) neurons releases kisspeptin into the median eminence, and neurokinin B (NKB) and dynorphin onto neighboring Kiss1ARH neurons to generate a slow EPSP mediated by TRPC5 channels that entrains intermittent, synchronous firing of Kiss1ARH neurons. High-frequency optogenetic stimulation of Kiss1ARH neurons also releases glutamate to excite the anorexigenic proopiomelanocortin (POMC) neurons and inhibit the orexigenic neuropeptide Y/agouti-related peptide (AgRP) neurons via metabotropic glutamate receptors. At the molecular level, the endoplasmic reticulum (ER) calcium-sensing protein stromal interaction molecule 1 (STIM1) is critically involved in the regulation of neuronal Ca2+ signaling and neuronal excitability through its interaction with plasma membrane (PM) calcium (e.g., TRPC) channels. Therefore, we hypothesized that deletion of Stim1 in Kiss1ARH neurons would increase neuronal excitability and their synchronous firing, which ultimately would affect energy homeostasis. Using optogenetics in combination with whole-cell recording and GCaMP6 imaging in slices, we discovered that deletion of Stim1 in Kiss1 neurons significantly increased the amplitude and duration of the slow EPSP and augmented synchronous [Ca2+]i oscillations in Kiss1ARH neurons. Deletion of Stim1 in Kiss1ARH neurons amplified the actions of NKB and protected ovariectomized female mice from developing obesity and glucose intolerance with high-fat dieting (HFD). Therefore, STIM1 appears to play a critical role in regulating synchronous firing of Kiss1ARH neurons, which ultimately affects the coordination between energy homeostasis and reproduction.SIGNIFICANCE STATEMENT Hypothalamic arcuate kisspeptin (Kiss1ARH) neurons are essential for stimulating the pulsatile release of gonadotropin-releasing hormone (GnRH) and maintaining fertility. However, Kiss1ARH neurons appear to be a key player in coordinating energy balance with reproduction. The regulation of calcium channels and hence calcium signaling is critically dependent on the endoplasmic reticulum (ER) calcium-sensing protein stromal interaction molecule 1 (STIM1), which interacts with the plasma membrane (PM) calcium channels. We have conditionally deleted Stim1 in Kiss1ARH neurons and found that it significantly increased the excitability of Kiss1ARH neurons and protected ovariectomized female mice from developing obesity and glucose intolerance with high-fat dieting (HFD).
Collapse
|
19
|
Vazquez MJ, Daza-Dueñas S, Tena-Sempere M. Emerging Roles of Epigenetics in the Control of Reproductive Function: Focus on Central Neuroendocrine Mechanisms. J Endocr Soc 2021; 5:bvab152. [PMID: 34703958 PMCID: PMC8533971 DOI: 10.1210/jendso/bvab152] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Indexed: 12/11/2022] Open
Abstract
Reproduction is an essential function for perpetuation of the species. As such, it is controlled by sophisticated regulatory mechanisms that allow a perfect match between environmental conditions and internal cues to ensure adequate pubertal maturation and achievement of reproductive capacity. Besides classical genetic regulatory events, mounting evidence has documented that different epigenetic mechanisms operate at different levels of the reproductive axis to finely tune the development and function of this complex neuroendocrine system along the lifespan. In this mini-review, we summarize recent evidence on the role of epigenetics in the control of reproduction, with special focus on the modulation of the central components of this axis. Particular attention will be paid to the epigenetic control of puberty and Kiss1 neurons because major developments have taken place in this domain recently. In addition, the putative role of central epigenetic mechanisms in mediating the influence of nutritional and environmental cues on reproductive function will be discussed.
Collapse
Affiliation(s)
- Maria Jesus Vazquez
- Instituto Maimónides de Investigación Biomédica de Cordoba (IMIBIC), 14004 Cordoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain.,Hospital Universitario Reina Sofia, 14004 Cordoba, Spain
| | - Silvia Daza-Dueñas
- Instituto Maimónides de Investigación Biomédica de Cordoba (IMIBIC), 14004 Cordoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain
| | - Manuel Tena-Sempere
- Instituto Maimónides de Investigación Biomédica de Cordoba (IMIBIC), 14004 Cordoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain.,Hospital Universitario Reina Sofia, 14004 Cordoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 14004 Cordoba, Spain.,Institute of Biomedicine, University of Turku, FIN-20520 Turku, Finland
| |
Collapse
|
20
|
Wang Z, Wu W, Kim MS, Cai D. GnRH pulse frequency and irregularity play a role in male aging. NATURE AGING 2021; 1:904-918. [PMID: 37118330 DOI: 10.1038/s43587-021-00116-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 08/12/2021] [Indexed: 04/30/2023]
Abstract
Gonadotropin-releasing hormone (GnRH) has a role in hypothalamic control of aging, but the underlying patterns and relationship with downstream reproductive hormones are still unclear. Here we report that hypothalamic GnRH pulse frequency and irregularity increase before GnRH pulse amplitude slowly decreases during aging. GnRH is inhibited by nuclear factor (NF)-κB, and GnRH pulses were controlled by oscillations in the transcriptional activity of NF-κB. Exposure to testosterone under pro-inflammatory conditions stimulated both NF-κB oscillations and GnRH pulses. While castration of middle-aged mice induced short-term anti-aging effects, preventing elevation of luteinizing hormone (LH) levels after castration led to long-term anti-aging effects and lifespan extension, indicating that high-frequency GnRH pulses and high-magnitude LH levels coordinately mediate aging. Reprogramming the endogenous GnRH pulses of middle-aged male mice via an optogenetic approach revealed that increasing GnRH pulses frequency causes LH excess and aging acceleration, while lowering the frequency of and stabilizing GnRH pulses can slow down aging. In conclusion, GnRH pulses are important for aging in male mice.
Collapse
Affiliation(s)
- Zhouguang Wang
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Wenhe Wu
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Min Soo Kim
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Dongsheng Cai
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
21
|
Dai M, Nakamura S, Takahashi C, Sato M, Munetomo A, Magata F, Uenoyama Y, Tsukamura H, Matsuda F. Reduction of arcuate kappa-opioid receptor-expressing cells increased luteinizing hormone pulse frequency in female rats. Endocr J 2021; 68:933-941. [PMID: 33867395 DOI: 10.1507/endocrj.ej20-0832] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The brain mechanism responsible for the pulsatile secretion of gonadotropin-releasing hormone (GnRH) is important for maintaining reproductive function in mammals. Accumulating evidence suggests that kisspeptin/neurokinin B/dynorphin A (KNDy) neurons in the hypothalamic arcuate nucleus (ARC) play a critical role in the regulation of pulsatile GnRH and subsequent gonadotropin secretion. Dynorphin A (Dyn) and its receptor, kappa-opioid receptor (KOR, encoded by Oprk1), have been shown to be involved in the suppression of pulsatile GnRH/luteinizing hormone (LH) release. On the other hand, it is still unclear whether the inhibitory Dyn signaling affects KNDy neurons or KOR-expressing non-KNDy cells in the ARC or other brain regions. We therefore aimed to clarify the role of ARC-specific Dyn-KOR signaling in the regulation of pulsatile GnRH/LH release by the ARC specific cell deletion of KOR-expressing cells using Dyn-conjugated-saporin (Dyn-SAP). Estrogen-primed ovariectomized female rats were administered Dyn-SAP to the ARC. In situ hybridization of Oprk1 showed that ARC Dyn-SAP administration significantly decreased the number of Oprk1-expressing cells in the ARC, but not in the ventromedial hypothalamic nucleus and paraventricular nucleus. The frequency of LH pulses significantly increased in animals bearing the ARC Dyn-SAP administration. The number of Kiss1-expressing cells in the ARC was not affected by ARC Dyn-SAP treatment. Dyn-KOR signaling within the ARC seems to mediate the suppression of the frequency of pulsatile GnRH/LH release, and ARC non-KNDy KOR neurons may be involved in the mechanism modulating GnRH/LH pulse generation.
Collapse
Affiliation(s)
- Mingdao Dai
- Laboratory of Theriogenology, Department of Veterinary Medical Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Sho Nakamura
- Laboratory of Theriogenology, Department of Veterinary Medical Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Chudai Takahashi
- Laboratory of Theriogenology, Department of Veterinary Medical Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Marimo Sato
- Laboratory of Theriogenology, Department of Veterinary Medical Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Arisa Munetomo
- Laboratory of Theriogenology, Department of Veterinary Medical Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Fumie Magata
- Laboratory of Theriogenology, Department of Veterinary Medical Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Yoshihisa Uenoyama
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Hiroko Tsukamura
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Fuko Matsuda
- Laboratory of Theriogenology, Department of Veterinary Medical Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| |
Collapse
|
22
|
Uenoyama Y, Inoue N, Nakamura S, Tsukamura H. Kisspeptin Neurons and Estrogen-Estrogen Receptor α Signaling: Unraveling the Mystery of Steroid Feedback System Regulating Mammalian Reproduction. Int J Mol Sci 2021; 22:ijms22179229. [PMID: 34502135 PMCID: PMC8430864 DOI: 10.3390/ijms22179229] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/21/2021] [Accepted: 08/23/2021] [Indexed: 12/13/2022] Open
Abstract
Estrogen produced by ovarian follicles plays a key role in the central mechanisms controlling reproduction via regulation of gonadotropin-releasing hormone (GnRH) release by its negative and positive feedback actions in female mammals. It has been well accepted that estrogen receptor α (ERα) mediates both estrogen feedback actions, but precise targets had remained as a mystery for decades. Ever since the discovery of kisspeptin neurons as afferent ERα-expressing neurons to govern GnRH neurons, the mechanisms mediating estrogen feedback are gradually being unraveled. The present article overviews the role of kisspeptin neurons in the arcuate nucleus (ARC), which are considered to drive pulsatile GnRH/gonadotropin release and folliculogenesis, in mediating the estrogen negative feedback action, and the role of kisspeptin neurons located in the anteroventral periventricular nucleus-periventricular nucleus (AVPV-PeN), which are thought to drive GnRH/luteinizing hormone (LH) surge and consequent ovulation, in mediating the estrogen positive feedback action. This implication has been confirmed by the studies showing that estrogen-bound ERα down- and up-regulates kisspeptin gene (Kiss1) expression in the ARC and AVPV-PeN kisspeptin neurons, respectively. The article also provides the molecular and epigenetic mechanisms regulating Kiss1 expression in kisspeptin neurons by estrogen. Further, afferent ERα-expressing neurons that may regulate kisspeptin release are discussed.
Collapse
Affiliation(s)
- Yoshihisa Uenoyama
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan; (Y.U.); (N.I.)
| | - Naoko Inoue
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan; (Y.U.); (N.I.)
| | - Sho Nakamura
- Faculty of Veterinary Medicine, Okayama University of Science, Imabari 794-8555, Japan;
| | - Hiroko Tsukamura
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan; (Y.U.); (N.I.)
- Correspondence:
| |
Collapse
|
23
|
Direct evidence that KNDy neurons maintain gonadotropin pulses and folliculogenesis as the GnRH pulse generator. Proc Natl Acad Sci U S A 2021; 118:2009156118. [PMID: 33500349 DOI: 10.1073/pnas.2009156118] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The gonadotropin-releasing hormone (GnRH) pulse is fundamental for mammalian reproduction: GnRH pulse regimens are needed as therapies for infertile women as continuous GnRH treatment paradoxically inhibits gonadotropin release. Circumstantial evidence suggests that the hypothalamic arcuate KNDy neurons expressing kisspeptin (encoded by Kiss1), neurokinin B (encoded by Tac3), and d ynorphin A serve as a GnRH pulse generator; however, no direct evidence is currently available. Here, we show that rescuing >20% KNDy neurons by transfecting Kiss1 inside arcuate Tac3 neurons, but not outside of these neurons, recovered folliculogenesis and luteinizing hormone (LH) pulses, an indicator of GnRH pulses, in female global Kiss1 knockout (KO) rats and that >90% conditional arcuate Kiss1 KO in newly generated Kiss1-floxed rats completely suppressed LH pulses. These results first provide direct evidence that KNDy neurons are the GnRH pulse generator, and at least 20% of KNDy neurons are sufficient to maintain folliculogenesis via generating GnRH/gonadotropin pulses.
Collapse
|
24
|
Stincic TL, Rønnekleiv OK, Kelly MJ. Membrane and nuclear initiated estrogenic regulation of homeostasis. Steroids 2021; 168:108428. [PMID: 31229508 PMCID: PMC6923613 DOI: 10.1016/j.steroids.2019.108428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 06/08/2019] [Accepted: 06/18/2019] [Indexed: 11/23/2022]
Abstract
Reproduction and energy balance are inextricably linked in order to optimize the evolutionary fitness of an organism. With insufficient or excessive energy stores a female is liable to suffer complications during pregnancy and produce unhealthy or obesity-prone offspring. The quintessential function of the hypothalamus is to act as a bridge between the endocrine and nervous systems, coordinating fertility and autonomic functions. Across the female reproductive cycle various motivations wax and wane, following levels of ovarian hormones. Estrogens, more specifically 17β-estradiol (E2), coordinate a triumvirate of hypothalamic neurons within the arcuate nucleus (ARH) that govern the physiological underpinnings of these behavioral dynamics. Arising from a common progenitor pool of cells, this triumvirate is composed of the kisspeptin (Kiss1ARH), proopiomelanocortin (POMC), and neuropeptide Y/agouti-related peptide (AgRP) neurons. Although the excitability of these neuronal subpopulations is subject to genomic and rapid estrogenic regulation, kisspeptin neurons are the most sensitive, reflecting their integral function in female fertility. Based on the premise that E2 coordinates autonomic functions around reproduction, we will review the recent findings on the synaptic interactions between Kiss1, AgRP and POMC neurons and how the rapid membrane-initiated and intracellular signaling cascades activated by E2 in these neurons are critical for control of homeostatic functions supporting reproduction.
Collapse
Affiliation(s)
- Todd L Stincic
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97239, United States
| | - Oline K Rønnekleiv
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97239, United States; Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, United States
| | - Martin J Kelly
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97239, United States; Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, United States.
| |
Collapse
|
25
|
Ruohonen ST, Poutanen M, Tena-Sempere M. Role of kisspeptins in the control of the hypothalamic-pituitary-ovarian axis: old dogmas and new challenges. Fertil Steril 2020; 114:465-474. [PMID: 32771258 DOI: 10.1016/j.fertnstert.2020.06.038] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 12/11/2022]
Abstract
In humans and other mammals, a hallmark of female reproductive function is the capacity to episodically release fertilizable oocytes under the precise control of a cascade of hormonal regulators that interplay in a cyclic manner within the hypothalamic-pituitary-ovarian (HPO) axis. Although the basic elements of this neurohormonal system were disclosed several decades before, a major breakthrough in our understanding of how the HPO axis is controlled during the lifespan came in the first decade of the 21st century, when the reproductive dimension of kisspeptins was disclosed by seminal studies documenting that genetic inactivation of the kisspeptin pathway is linked to central hypogonadism and infertility. Kisspeptins are a family of peptides, encoded by the Kiss1 gene, that operate via the surface receptor, Gpr54 (also called Kiss1r), to regulate virtually all aspects of reproduction in both sexes. The primary site of action of kisspeptins is the hypothalamus, where Kiss1 neurons engage in the precise control of the pulsatile release of GnRH to modulate gonadotropin secretion and, thereby, ovarian function. Nonetheless, additional sites of action of kisspeptins within the HPO axis, including the pituitary and the ovary, have been proposed; yet, the physiologic relevance of such extrahypothalamic actions of kisspeptins is still a matter of debate. In this review, we summarize the current consensus knowledge and open questions on the sites of action, physiologic roles, and eventual therapeutic implications of kisspeptins in the control of the female reproductive axis.
Collapse
Affiliation(s)
- Suvi T Ruohonen
- Research Center for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland; Turku Center for Disease Modeling, Turku, Finland
| | - Matti Poutanen
- Research Center for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland; Turku Center for Disease Modeling, Turku, Finland
| | - Manuel Tena-Sempere
- Research Center for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland; Turku Center for Disease Modeling, Turku, Finland; Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain; Instituto Maimónides de Investigación Biomédica de Córdoba and Hospital Universitario Reina Sofia, Córdoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Córdoba, Spain.
| |
Collapse
|
26
|
Kem DC, Li H, Yu X, Weedin E, Reynolds AC, Forsythe E, Beel M, Fischer H, Hines B, Guo Y, Deng J, Liles JT, Nuss Z, Elkosseifi M, Aston CE, Burks HR, Craig LB. The Role of GnRH Receptor Autoantibodies in Polycystic Ovary Syndrome. J Endocr Soc 2020; 4:bvaa078. [PMID: 32803090 PMCID: PMC7417878 DOI: 10.1210/jendso/bvaa078] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/11/2020] [Indexed: 12/28/2022] Open
Abstract
Objective Is polycystic ovary syndrome (PCOS) associated with activating autoantibodies (AAb) to the second extracellular loop (ECL2) of gonadotropin-releasing hormone receptor (GnRHR)? Design and Methods We retrospectively screened sera from 40 patients with PCOS and 14 normal controls (NCs) with regular menses using enzyme-linked immunosorbent assay (ELISA) for the presence of GnRHR-ECL2-AAb. We obtained similar data from 40 non-PCOS ovulatory but infertile patients as a control group (OIC) of interest. We analyzed GnRHR-ECL2-AAb activity in purified immunoglobulin (Ig)G using a cell-based GnRHR bioassay. Results The mean ELISA value in the PCOS group was markedly higher than the NC (P = .000036) and the OIC (P = .0028) groups. IgG from a sample of 5 PCOS subjects, in contrast to a sample of 5 OIC subjects, demonstrated a dose-dependent increase in GnRHR-stimulating activity qualitatively similar to the acute action of the natural ligand GnRH and the synthetic agonist leuprolide. The GnRHR antagonist cetrorelix significantly suppressed (P < .01) the elevated GnRHR activity induced by IgG from 7 PCOS patients while the IgG activity level from 7 OIC subjects was unchanged. Five other OIC subjects had relatively high ELISA values at or above the 95% confidence limits. On further study, 3 had normal or low activity while 2 had elevated IgG-induced GnRHR activity. One suppressed with cetrorelix while the other did not. The copresence of PCOS IgG increased the responsiveness to GnRH and shifted the dosage response curve to the left (P < .01). Conclusions GnRHR-ECL2-AAb are significantly elevated in patients with PCOS compared with NCs. Their presence raises important etiological, diagnostic, and therapeutic implications.
Collapse
Affiliation(s)
- David C Kem
- Section of Endocrinology and Diabetes, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma city, Oklahoma.,VA Medical Center, Oklahoma City, Oklahoma
| | - Hongliang Li
- Section of Endocrinology and Diabetes, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma city, Oklahoma
| | - Xichun Yu
- Section of Endocrinology and Diabetes, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma city, Oklahoma
| | - Elizabeth Weedin
- Section of Reproductive Endocrinology & Infertility, Department of Obstetrics & Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Anna C Reynolds
- Section of Reproductive Endocrinology & Infertility, Department of Obstetrics & Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Elizabeth Forsythe
- Section of Endocrinology and Diabetes, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma city, Oklahoma
| | - Marci Beel
- Section of Endocrinology and Diabetes, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma city, Oklahoma
| | - Hayley Fischer
- Section of Endocrinology and Diabetes, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma city, Oklahoma
| | - Brendon Hines
- Section of Endocrinology and Diabetes, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma city, Oklahoma
| | - Yankai Guo
- Section of Endocrinology and Diabetes, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma city, Oklahoma
| | - Jielin Deng
- Section of Endocrinology and Diabetes, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma city, Oklahoma
| | - Jonathan T Liles
- Section of Endocrinology and Diabetes, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma city, Oklahoma
| | - Zachary Nuss
- Section of Endocrinology and Diabetes, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma city, Oklahoma
| | - Myriam Elkosseifi
- Section of Endocrinology and Diabetes, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma city, Oklahoma
| | - Christopher E Aston
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Heather R Burks
- Section of Reproductive Endocrinology & Infertility, Department of Obstetrics & Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - LaTasha B Craig
- Section of Reproductive Endocrinology & Infertility, Department of Obstetrics & Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
27
|
Ikegami K, Goto T, Nakamura S, Watanabe Y, Sugimoto A, Majarune S, Horihata K, Nagae M, Tomikawa J, Imamura T, Sanbo M, Hirabayashi M, Inoue N, Maeda KI, Tsukamura H, Uenoyama Y. Conditional kisspeptin neuron-specific Kiss1 knockout with newly generated Kiss1-floxed and Kiss1-Cre mice replicates a hypogonadal phenotype of global Kiss1 knockout mice. J Reprod Dev 2020; 66:359-367. [PMID: 32307336 PMCID: PMC7470906 DOI: 10.1262/jrd.2020-026] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The present study aimed to evaluate whether novel conditional kisspeptin neuron-specific Kiss1 knockout (KO) mice utilizing the Cre-loxP system could recapitulate
the infertility of global Kiss1 KO models, thereby providing further evidence for the fundamental role of hypothalamic kisspeptin neurons in regulating mammalian
reproduction. We generated Kiss1-floxed mice and hypothalamic kisspeptin neuron-specific Cre-expressing transgenic mice and then crossed these two
lines. The conditional Kiss1 KO mice showed pubertal failure along with a suppression of gonadotropin secretion and ovarian atrophy. These results indicate that
newly-created hypothalamic Kiss1 KO mice obtained by the Cre-loxP system recapitulated the infertility of global Kiss1 KO models, suggesting that
hypothalamic kisspeptin, but not peripheral kisspeptin, is critical for reproduction. Importantly, these Kiss1-floxed mice are now available and will be a valuable
tool for detailed analyses of roles of each population of kisspeptin neurons in the brain and peripheral kisspeptin-producing cells by the spatiotemporal-specific manipulation of
Cre expression.
Collapse
Affiliation(s)
- Kana Ikegami
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Teppei Goto
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan.,Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
| | - Sho Nakamura
- Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Youki Watanabe
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Arisa Sugimoto
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Sutisa Majarune
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Kei Horihata
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Mayuko Nagae
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Junko Tomikawa
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Takuya Imamura
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8526, Japan
| | - Makoto Sanbo
- Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
| | - Masumi Hirabayashi
- Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
| | - Naoko Inoue
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Kei-Ichiro Maeda
- Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Hiroko Tsukamura
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Yoshihisa Uenoyama
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| |
Collapse
|
28
|
Si W, Li H, Kang T, Ye J, Yao Z, Liu Y, Yu T, Zhang Y, Ling Y, Cao H, Wang J, Li Y, Fang F. Effect of GABA-T on Reproductive Function in Female Rats. Animals (Basel) 2020; 10:ani10040567. [PMID: 32230949 PMCID: PMC7222393 DOI: 10.3390/ani10040567] [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: 02/08/2020] [Revised: 03/16/2020] [Accepted: 03/25/2020] [Indexed: 12/29/2022] Open
Abstract
This study explored the role of γ-aminobutyric acid transaminase (GABA-T) in the puberty and reproductive performance of female rats. Immunofluorescence technique, quantitative real-time PCR (RT-qPCR) and enzyme-linked immunosorbent assay (ELISA) were used to detect the distribution of GABA-T and the expression of genes and hormones in female rats, respectively. The results showed that GABA-T was mainly distributed in the arcuate nucleus (ARC), paraventricular nucleus (PVN) and periventricular nucleus (PeN) of the hypothalamus, and in the adenohypophysis, ovarian granulosa cells and oocytes. Abat mRNA level at 28 d was lowest in the hypothalamus and the pituitary; at puberty, it was lowest in the ovary. Abat mRNA level was highest in adults in the hypothalamus; at infancy and puberty, it was highest in the pituitary; and at 21 d it was highest in the ovary. After vigabatrin (GABA-T irreversible inhibitor) was added to hypothalamus cells, the levels of Abat mRNA and Rfrp-3 mRNA were significantly reduced, but Gnrh mRNA increased at the dose of 25 and 50 μg/mL; Kiss1 mRNA was significantly increased but Gabbr1 mRNA was reduced at the 50 μg/mL dose. In prepubertal rats injected with vigabatrin, puberty onset was delayed. Abat mRNA, Kiss1 mRNA and Gnrh mRNA levels were significantly reduced, but Rfrp-3 mRNA level increased in the hypothalamus. Vigabatrin reduced the concentrations of GABA-T, luteinizing hormone (LH) and progesterone (P4), and the ovarian index. Lactation performance was reduced in adult rats with vigabatrin treatment. Four hours after vigabatrin injection, the concentrations of GABA-T and LH were significantly reduced in adult and 25 d rats, but follicle-stimulating hormone (FSH) increased in 25 d rats. In conclusion, GABA-T affects the reproductive function of female rats by regulating the levels of Gnrh, Kiss1 and Rfrp-3 in the hypothalamus as well as the concentrations of LH and P4.
Collapse
Affiliation(s)
- Wenyu Si
- Anhui Provincial Laboratory of Animal Genetic Resources Protection and Breeding, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei 230036, China; (W.S.); (H.L.); (T.K.); (J.Y.); (Z.Y.); (Y.L.); (T.Y.); (Y.Z.); (Y.L.); (H.C.); (J.W.); (Y.L.)
- Anhui Provincial Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, 130 Changjiang West Road, Hefei 230036, China
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei 230036, China
| | - Hailing Li
- Anhui Provincial Laboratory of Animal Genetic Resources Protection and Breeding, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei 230036, China; (W.S.); (H.L.); (T.K.); (J.Y.); (Z.Y.); (Y.L.); (T.Y.); (Y.Z.); (Y.L.); (H.C.); (J.W.); (Y.L.)
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei 230036, China
| | - Tiezhu Kang
- Anhui Provincial Laboratory of Animal Genetic Resources Protection and Breeding, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei 230036, China; (W.S.); (H.L.); (T.K.); (J.Y.); (Z.Y.); (Y.L.); (T.Y.); (Y.Z.); (Y.L.); (H.C.); (J.W.); (Y.L.)
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei 230036, China
| | - Jing Ye
- Anhui Provincial Laboratory of Animal Genetic Resources Protection and Breeding, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei 230036, China; (W.S.); (H.L.); (T.K.); (J.Y.); (Z.Y.); (Y.L.); (T.Y.); (Y.Z.); (Y.L.); (H.C.); (J.W.); (Y.L.)
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei 230036, China
| | - Zhiqiu Yao
- Anhui Provincial Laboratory of Animal Genetic Resources Protection and Breeding, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei 230036, China; (W.S.); (H.L.); (T.K.); (J.Y.); (Z.Y.); (Y.L.); (T.Y.); (Y.Z.); (Y.L.); (H.C.); (J.W.); (Y.L.)
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei 230036, China
| | - Ya Liu
- Anhui Provincial Laboratory of Animal Genetic Resources Protection and Breeding, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei 230036, China; (W.S.); (H.L.); (T.K.); (J.Y.); (Z.Y.); (Y.L.); (T.Y.); (Y.Z.); (Y.L.); (H.C.); (J.W.); (Y.L.)
- Anhui Provincial Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, 130 Changjiang West Road, Hefei 230036, China
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei 230036, China
| | - Tong Yu
- Anhui Provincial Laboratory of Animal Genetic Resources Protection and Breeding, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei 230036, China; (W.S.); (H.L.); (T.K.); (J.Y.); (Z.Y.); (Y.L.); (T.Y.); (Y.Z.); (Y.L.); (H.C.); (J.W.); (Y.L.)
- Anhui Provincial Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, 130 Changjiang West Road, Hefei 230036, China
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei 230036, China
| | - Yunhai Zhang
- Anhui Provincial Laboratory of Animal Genetic Resources Protection and Breeding, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei 230036, China; (W.S.); (H.L.); (T.K.); (J.Y.); (Z.Y.); (Y.L.); (T.Y.); (Y.Z.); (Y.L.); (H.C.); (J.W.); (Y.L.)
- Anhui Provincial Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, 130 Changjiang West Road, Hefei 230036, China
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei 230036, China
| | - Yinghui Ling
- Anhui Provincial Laboratory of Animal Genetic Resources Protection and Breeding, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei 230036, China; (W.S.); (H.L.); (T.K.); (J.Y.); (Z.Y.); (Y.L.); (T.Y.); (Y.Z.); (Y.L.); (H.C.); (J.W.); (Y.L.)
- Anhui Provincial Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, 130 Changjiang West Road, Hefei 230036, China
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei 230036, China
| | - Hongguo Cao
- Anhui Provincial Laboratory of Animal Genetic Resources Protection and Breeding, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei 230036, China; (W.S.); (H.L.); (T.K.); (J.Y.); (Z.Y.); (Y.L.); (T.Y.); (Y.Z.); (Y.L.); (H.C.); (J.W.); (Y.L.)
- Anhui Provincial Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, 130 Changjiang West Road, Hefei 230036, China
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei 230036, China
| | - Juhua Wang
- Anhui Provincial Laboratory of Animal Genetic Resources Protection and Breeding, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei 230036, China; (W.S.); (H.L.); (T.K.); (J.Y.); (Z.Y.); (Y.L.); (T.Y.); (Y.Z.); (Y.L.); (H.C.); (J.W.); (Y.L.)
- Anhui Provincial Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, 130 Changjiang West Road, Hefei 230036, China
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei 230036, China
| | - Yunsheng Li
- Anhui Provincial Laboratory of Animal Genetic Resources Protection and Breeding, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei 230036, China; (W.S.); (H.L.); (T.K.); (J.Y.); (Z.Y.); (Y.L.); (T.Y.); (Y.Z.); (Y.L.); (H.C.); (J.W.); (Y.L.)
- Anhui Provincial Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, 130 Changjiang West Road, Hefei 230036, China
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei 230036, China
| | - Fugui Fang
- Anhui Provincial Laboratory of Animal Genetic Resources Protection and Breeding, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei 230036, China; (W.S.); (H.L.); (T.K.); (J.Y.); (Z.Y.); (Y.L.); (T.Y.); (Y.Z.); (Y.L.); (H.C.); (J.W.); (Y.L.)
- Anhui Provincial Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, 130 Changjiang West Road, Hefei 230036, China
- Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei 230036, China
- Correspondence:
| |
Collapse
|
29
|
Ullah A, Pirzada M, Jahan S, Ullah H, Razak S, Rauf N, Khan MJ, Mahboob SZ. Prenatal BPA and its analogs BPB, BPF, and BPS exposure and reproductive axis function in the male offspring of Sprague Dawley rats. Hum Exp Toxicol 2020; 38:1344-1365. [PMID: 31514588 DOI: 10.1177/0960327119862335] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Research in the past has indicated associated long-term and low levels of exposure of bisphenol A (BPA) in early life and neuroendocrine disorders, such as obesity, precocious puberty, diabetes, and hypertension. BPA and its analogs bisphenol B (BPB), bisphenol F (BPF), and bisphenol S (BPS) have been reported to have similar or even more toxic effect as compared to BPA. Exposure of rats to BPA and its analogs BPB, BPF, and BPS resulted in decreased sperm production, testosterone secretion, and histological changes in the reproductive tissues of male rats. In the present study, BPA, BPB, BPF, and BPS were administered in drinking water at concentrations of (5, 25, and 50 μg/L) from pregnancy day (PD) 1 to PD 21. Body weight (BW), hormonal concentrations, antioxidant enzymes, and histological changes were determined in the reproductive tissues. BPA and its analogs prenatal exposure to female rats induced significant statistical difference in the antioxidant enzymes, plasma testosterone, and estrogen concentrations in the male offspring when compared with the control. Histological parameters of both testis and epididymis revealed prominent changes in the reproductive tissues. The present study suggests that BPA and its analogs BPB, BPF, and BPS different concentrations led to marked alterations in the development of the male reproductive system.
Collapse
Affiliation(s)
- A Ullah
- Reproductive Physiology Laboratory, Department of Animal Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - M Pirzada
- Reproductive Physiology Laboratory, Department of Animal Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - S Jahan
- Reproductive Physiology Laboratory, Department of Animal Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - H Ullah
- Reproductive Physiology Laboratory, Department of Animal Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - S Razak
- Reproductive Physiology Laboratory, Department of Animal Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - N Rauf
- Reproductive Physiology Laboratory, Department of Animal Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - M J Khan
- Reproductive Physiology Laboratory, Department of Animal Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - S Z Mahboob
- Reproductive Physiology Laboratory, Department of Animal Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| |
Collapse
|
30
|
Kanda S. Evolution of the regulatory mechanisms for the hypothalamic-pituitary-gonadal axis in vertebrates-hypothesis from a comparative view. Gen Comp Endocrinol 2019; 284:113075. [PMID: 30500374 DOI: 10.1016/j.ygcen.2018.11.014] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 11/26/2018] [Accepted: 11/26/2018] [Indexed: 12/26/2022]
Abstract
Reproduction is regulated by the hypothalamic-pituitary-gonadal (HPG) axis in vertebrates. In addition to wealth of knowledge in mammals, recent studies in non-mammalian species, especially teleosts, have provided evidence that some of the components in the HPG axis are conserved in bony vertebrates. On the other hand, from the comparisons of the recent accumulating knowledge between mammals and teleosts, unique characteristics of the regulatory system in each group have been unveiled. A hypophysiotropic neurotransmitter/hormone, gonadotropin releasing hormone (GnRH), pituitary gonadotropins, follicle stimulating hormone (FSH), and luteinizing hormone (LH) were proven to be common important elements of the HPG axis in teleosts and mammals, although the roles of each vary. Conversely, there are some modulators of GnRH or gonadotropins that are not common to all vertebrates. In this review, I will introduce the mechanism for HPG axis regulation in mammals and teleosts, and describe their evolution from a hypothetical common ancestor.
Collapse
Affiliation(s)
- Shinji Kanda
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo 113-0033, Japan.
| |
Collapse
|
31
|
Hawken PAR, Smith JT, Jorre de St Jorre T, Esmaili T, Scott CJ, Rodger J, Blache D, Martin GB. Patterns of preoptic-hypothalamic neuronal activation and LH secretion in female sheep following the introduction and withdrawal of novel males. Reprod Fertil Dev 2019; 31:1674-1681. [PMID: 31511142 DOI: 10.1071/rd19079] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 06/14/2019] [Indexed: 11/23/2022] Open
Abstract
The neuroendocrine response of female sheep to a novel male involves neural activation in the hypothalamus. However, if males are removed, the gonadotrophic signal declines, so the neural activity is likely to change. We examined Fos-immunoreactive (IR) cells in hypothalamic tissues from seasonally anovulatory female sheep exposed to males for 2 or 6h, or for 2h followed by 4h isolation from males. Control females were killed in the absence of male exposure. Male introduction increased LH secretion in all females; male removal was associated with a reduction only in mean and basal LH concentrations. Females exposed to males for 2h had more Fos-IR cells in the arcuate nucleus (ARC), ventromedial nucleus of the hypothalamus (VMH) and organum vasculosum of the lamina terminalis (OVLT) than control females. Fos-IR cells in the preoptic area (POA) were only greater than in control females after 6h exposure to a male. Removal of males decreased the number of Fos-IR cells in the ARC, VMH and OVLT, but not in the POA. Thus, hypothalamic neural activation and LH secretion in female sheep are stimulated by males and decline after male removal. However, activation in the POA persists after removal and may explain the incomplete decline in the LH response.
Collapse
Affiliation(s)
- Penny A R Hawken
- School of Agriculture and Environment, The University of Western Australia, Crawley, WA 6009, Australia
| | - Jeremy T Smith
- School of Human Sciences, The University of Western Australia, Crawley, WA 6009, Australia
| | - Trina Jorre de St Jorre
- School of Agriculture and Environment, The University of Western Australia, Crawley, WA 6009, Australia
| | - Tammi Esmaili
- La Trobe University, Cnr Plenty Road and Kingsbury Drive, Melbourne, Vic. 3086, Australia
| | - Christopher J Scott
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, NSW 2678, Australia
| | - Jennifer Rodger
- School of Biological Sciences, The University of Western Australia, Crawley, WA 6009, Australia; and Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| | - Dominique Blache
- School of Agriculture and Environment, The University of Western Australia, Crawley, WA 6009, Australia
| | - Graeme B Martin
- School of Agriculture and Environment, The University of Western Australia, Crawley, WA 6009, Australia; and Corresponding author.
| |
Collapse
|
32
|
Majarune S, Nima P, Sugimoto A, Nagae M, Inoue N, Tsukamura H, Uenoyama Y. Ad libitum feeding triggers puberty onset associated with increases in arcuate Kiss1 and Pdyn expression in growth-retarded rats. J Reprod Dev 2019; 65:397-406. [PMID: 31155522 PMCID: PMC6815743 DOI: 10.1262/jrd.2019-048] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Increasing evidence shows that puberty onset is largely dependent on body weight rather than chronological age. To investigate the mechanism involved in the energetic control of puberty
onset, the present study examined effects of chronic food restriction during the prepubertal period and the resumption of ad libitum feeding for 24 and 48 h on estrous
cyclicity, Kiss1 (kisspeptin gene), Tac3 (neurokinin B gene) and Pdyn (dynorphin A gene) expression in the hypothalamus, luteinizing
hormone (LH) secretion and follicular development in female rats. When animals weighed 75 g, they were subjected to a restricted feeding to retard growth to 70–80 g by 49 days of age. Then,
animals were subjected to ad libitum feeding or remained food-restricted. The growth-retarded rats did not show puberty onset associated with suppression of both
Kiss1 and Pdyn expression in the arcuate nucleus (ARC). 24-h ad libitum feeding increased tonic LH secretion and the number of Graafian
and non-Graafian tertiary follicles with an increase in the numbers of ARC Kiss1- and Pdyn-expressing cells. 48-h ad libitum feeding
induced the vaginal proestrus and a surge-like LH increase with an increase in Kiss1-expressing cells in the anteroventral periventricular nucleus (AVPV). These results
suggest that the negative energy balance causes pubertal failure with suppression of ARC Kiss1 and Pdyn expression and then subsequent gonadotropin
secretion and ovarian function, while the positive energetic cues trigger puberty onset via an increase in ARC Kiss1 and Pdyn expression and thus
gonadotropin secretion and follicular development in female rats.
Collapse
Affiliation(s)
- Sutisa Majarune
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Pelden Nima
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Arisa Sugimoto
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Mayuko Nagae
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Naoko Inoue
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Hiroko Tsukamura
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Yoshihisa Uenoyama
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| |
Collapse
|
33
|
Uenoyama Y, Inoue N, Maeda KI, Tsukamura H. The roles of kisspeptin in the mechanism underlying reproductive functions in mammals. J Reprod Dev 2018; 64:469-476. [PMID: 30298825 PMCID: PMC6305848 DOI: 10.1262/jrd.2018-110] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Kisspeptin, identified as a natural ligand of GPR54 in 2001, is now considered as a master regulator of puberty and subsequent reproductive functions in mammals. Our previous studies using
Kiss1 knockout (KO) rats clearly demonstrated the indispensable role of kisspeptin in gonadotropin-releasing hormone (GnRH)/gonadotropin secretion. In addition, behavioral
analyses of Kiss1 KO rats revealed an organizational effect of kisspeptin on neural circuits controlling sexual behaviors. Our studies using transgenic mice carrying a
region-specific Kiss1 enhancer-driven reporter gene provided a clue as to the mechanism by which estrogen regulates Kiss1 expression in hypothalamic
kisspeptin neurons. Analyses of Kiss1 expression and gonadotropin secretion during the pubertal transition shed light on the mechanism triggering GnRH/gonadotropin secretion
at the onset of puberty in rats. Here, we summarize data obtained from the aforementioned studies and revisit the physiological roles of kisspeptin in the mechanism underlying reproductive
functions in mammals.
Collapse
Affiliation(s)
- Yoshihisa Uenoyama
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Naoko Inoue
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Kei-Ichiro Maeda
- Department of Veterinary Medical Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Hiroko Tsukamura
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| |
Collapse
|
34
|
Nestor CC, Bedenbaugh MN, Hileman SM, Coolen LM, Lehman MN, Goodman RL. Regulation of GnRH pulsatility in ewes. Reproduction 2018; 156:R83-R99. [PMID: 29880718 DOI: 10.1530/rep-18-0127] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 06/07/2018] [Indexed: 01/21/2023]
Abstract
Early work in ewes provided a wealth of information on the physiological regulation of pulsatile gonadotropin-releasing hormone (GnRH) secretion by internal and external inputs. Identification of the neural systems involved, however, was limited by the lack of information on neural mechanisms underlying generation of GnRH pulses. Over the last decade, considerable evidence supported the hypothesis that a group of neurons in the arcuate nucleus that contain kisspeptin, neurokinin B and dynorphin (KNDy neurons) are responsible for synchronizing secretion of GnRH during each pulse in ewes. In this review, we describe our current understanding of the neural systems mediating the actions of ovarian steroids and three external inputs on GnRH pulsatility in light of the hypothesis that KNDy neurons play a key role in GnRH pulse generation. In breeding season adults, estradiol (E2) and progesterone decrease GnRH pulse amplitude and frequency, respectively, by actions on KNDy neurons, with E2 decreasing kisspeptin and progesterone increasing dynorphin release onto GnRH neurons. In pre-pubertal lambs, E2 inhibits GnRH pulse frequency by decreasing kisspeptin and increasing dynorphin release, actions that wane as the lamb matures to allow increased pulsatile GnRH secretion at puberty. Less is known about mediators of undernutrition and stress, although some evidence implicates kisspeptin and dynorphin, respectively, in the inhibition of GnRH pulse frequency by these factors. During the anoestrus, inhibitory photoperiod acting via melatonin activates A15 dopaminergic neurons that innervate KNDy neurons; E2 increases dopamine release from these neurons to inhibit KNDy neurons and suppress the frequency of kisspeptin and GnRH release.
Collapse
Affiliation(s)
- Casey C Nestor
- Department of Animal Science, North Carolina State University, Raleigh, North Carolina, USA
| | - Michelle N Bedenbaugh
- Department of Physiology, Pharmacology and Neuroscience, West Virginia University, Morgantown, West Virginia, USA
| | - Stanley M Hileman
- Department of Physiology, Pharmacology and Neuroscience, West Virginia University, Morgantown, West Virginia, USA
| | - Lique M Coolen
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA.,Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Michael N Lehman
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Robert L Goodman
- Department of Physiology, Pharmacology and Neuroscience, West Virginia University, Morgantown, West Virginia, USA
| |
Collapse
|
35
|
Franssen D, Tena-Sempere M. The kisspeptin receptor: A key G-protein-coupled receptor in the control of the reproductive axis. Best Pract Res Clin Endocrinol Metab 2018; 32:107-123. [PMID: 29678280 DOI: 10.1016/j.beem.2018.01.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The kisspeptin receptor, Kiss1R, also known as Gpr54, is a G protein-coupled receptor (GPCR), deorphanized in 2001, when it was recognized as canonical receptor for the Kiss1-derived peptides, kisspeptins. In 2003, inactivating mutations of Kiss1R gene were first associated to lack of pubertal maturation and hypogonadotropic hypogonadism in humans and rodents. These seminal findings pointed out the previously unsuspected, essential role of Kiss1R and its ligands in control of reproductive maturation and function. This contention has been fully substantiated during the last decade by a wealth of clinical and experimental data, which has documented a fundamental function of the so-called Kiss1/Kiss1R system in the regulation of puberty onset, gonadotropin secretion and ovulation, as well as the metabolic and environmental modulation of fertility. In this review, we provide a succinct summary of some of the most salient facets of Kiss1R, as essential GPCR for the proper maturation and function of the reproductive axis.
Collapse
Affiliation(s)
- Delphine Franssen
- Instituto Maimónides de Investigación Biomédica de Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004, Cordoba, Spain; Hospital Universitario Reina Sofia, 14004, Cordoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 14004, Cordoba, Spain
| | - Manuel Tena-Sempere
- Instituto Maimónides de Investigación Biomédica de Cordoba (IMIBIC), 14004, Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004, Cordoba, Spain; Hospital Universitario Reina Sofia, 14004, Cordoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 14004, Cordoba, Spain; FiDiPro Program, Institute of Biomedicine, University of Turku, FIN-20520, Turku, Finland.
| |
Collapse
|
36
|
Assadullah, Ieda N, Kawai N, Ishii H, Ihara K, Inoue N, Uenoyama Y, Tsukamura H. Co-expression of the calcitonin receptor gene in the hypothalamic kisspeptin neurons in female rats. Reprod Med Biol 2018; 17:164-172. [PMID: 29692674 PMCID: PMC5902471 DOI: 10.1002/rmb2.12085] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 12/20/2017] [Indexed: 11/25/2022] Open
Abstract
Purpose Hypothalamic kisspeptin neurons are considered to play a critical role in regulating mammalian reproduction and integrating humoral and neuronal inputs that control gonadotropin‐releasing hormone (GnRH)/gonadotropin release. The present study aimed to investigate the upstream regulator candidates for kisspeptin neurons. Methods Visualized kisspeptin neurons that were taken from the arcuate nucleus (ARC) of Kiss1‐tdTomato rats were subjected to next‐generation sequencing (NGS) analysis. In situ hybridization (ISH) for the calcitonin receptor gene (Calcr) was performed throughout the whole forebrain of ovariectomized wild‐type female rats that had been implanted with a negative feedback level of estrogen, because the Calcr expression was evident in the ARC kisspeptin neurons from the NGS analysis. Then, a double ISH was performed for the Calcr and kisspeptin gene (Kiss1) in the brain regions, containing either the anteroventral periventricular nucleus (AVPV) or ARC of the female rats. Results The NGS analysis revealed that the Calcr was highly expressed in the ARC kisspeptin neurons. It was found that the Calcr was co‐expressed in 12% and 22% of the Kiss1‐expressing cells in the ARC and AVPV, respectively. Conclusion The present study suggests that calcitonin receptor signaling could be involved in the regulation of reproductive function through the direct control of the ARC and/or AVPV kisspeptin neurons, and then GnRH/gonadotropin release.
Collapse
Affiliation(s)
- Assadullah
- Graduate School of Bioagricultural Sciences Nagoya University Nagoya Japan
| | - Nahoko Ieda
- Graduate School of Bioagricultural Sciences Nagoya University Nagoya Japan
| | - Narumi Kawai
- Graduate School of Bioagricultural Sciences Nagoya University Nagoya Japan
| | - Hirotaka Ishii
- Department of Anatomy and Neurobiology Nippon Medical School Tokyo Japan
| | - Kunio Ihara
- Center for Gene Research Nagoya University Nagoya Japan
| | - Naoko Inoue
- Graduate School of Bioagricultural Sciences Nagoya University Nagoya Japan
| | - Yoshihisa Uenoyama
- Graduate School of Bioagricultural Sciences Nagoya University Nagoya Japan
| | - Hiroko Tsukamura
- Graduate School of Bioagricultural Sciences Nagoya University Nagoya Japan
| |
Collapse
|
37
|
Weems PW, Lehman MN, Coolen LM, Goodman RL. The Roles of Neurokinins and Endogenous Opioid Peptides in Control of Pulsatile LH Secretion. VITAMINS AND HORMONES 2018; 107:89-135. [PMID: 29544644 DOI: 10.1016/bs.vh.2018.01.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
Abstract
Work over the last 15 years on the control of pulsatile LH secretion has focused largely on a set of neurons in the arcuate nucleus (ARC) that contains two stimulatory neuropeptides, critical for fertility in humans (kisspeptin and neurokinin B (NKB)) and the inhibitory endogenous opioid peptide (EOP), dynorphin, and are now known as KNDy (kisspeptin-NKB-dynorphin) neurons. In this review, we consider the role of each of the KNDy peptides in the generation of GnRH pulses and the negative feedback actions of ovarian steroids, with an emphasis on NKB and dynorphin. With regard to negative feedback, there appear to be important species differences. In sheep, progesterone inhibits GnRH pulse frequency by stimulating dynorphin release, and estradiol inhibits pulse amplitude by suppressing kisspeptin. In rodents, the role of KNDy neurons in estrogen negative feedback remains controversial, progesterone may inhibit GnRH via dynorphin, but the physiological significance of this action is unclear. In primates, an EOP, probably dynorphin, mediates progesterone negative feedback, and estrogen inhibits kisspeptin expression. In contrast, there is now compelling evidence from several species that kisspeptin is the output signal from KNDy neurons that drives GnRH release during a pulse and may also act within the KNDy network to affect pulse frequency. NKB is thought to act within this network to initiate each pulse, although there is some redundancy in tachykinin signaling in rodents. In ruminants, dynorphin terminates GnRH secretion at the end of pulse, most likely acting on both KNDy and GnRH neurons, but the data on the role of this EOP in rodents are conflicting.
Collapse
Affiliation(s)
- Peyton W Weems
- Graduate Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, United States
| | - Michael N Lehman
- University of Mississippi Medical Center, Jackson, MS, United States
| | - Lique M Coolen
- University of Mississippi Medical Center, Jackson, MS, United States
| | | |
Collapse
|
38
|
Takács S, Bardóczi Z, Skrapits K, Göcz B, Váczi V, Maglóczky Z, Szűcs I, Rácz G, Matolcsy A, Dhillo WS, Watanabe M, Kádár A, Fekete C, Kalló I, Hrabovszky E. Post mortem single-cell labeling with DiI and immunoelectron microscopy unveil the fine structure of kisspeptin neurons in humans. Brain Struct Funct 2018; 223:2143-2156. [PMID: 29380121 DOI: 10.1007/s00429-018-1610-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 01/10/2018] [Indexed: 11/30/2022]
Abstract
Kisspeptin (KP) synthesizing neurons of the hypothalamic infundibular region are critically involved in the central regulation of fertility; these cells regulate pulsatile gonadotropin-releasing hormone (GnRH) secretion and mediate sex steroid feedback signals to GnRH neurons. Fine structural analysis of the human KP system is complicated by the use of post mortem tissues. To gain better insight into the neuroanatomy of the somato-dendritic cellular compartment, we introduced the diolistic labeling of immunohistochemically identified KP neurons using a gene gun loaded with the lipophilic dye, DiI. Confocal microscopic studies of primary dendrites in 100-µm-thick tissue sections established that 79.3% of KP cells were bipolar, 14.1% were tripolar, and 6.6% were unipolar. Primary dendrites branched sparsely, contained numerous appendages (9.1 ± 1.1 spines/100 µm dendrite), and received rich innervation from GABAergic, glutamatergic, and KP-containing terminals. KP neuron synaptology was analyzed with immunoelectron microscopy on perfusion-fixed specimens. KP axons established frequent contacts and classical synapses on unlabeled, and on KP-immunoreactive somata, dendrites, and spines. Synapses were asymmetric and the presynaptic structures contained round and regular synaptic vesicles, in addition to dense-core granules. Although immunofluorescent studies failed to detect vesicular glutamate transporter isoforms in KP axons, ultrastructural characteristics of synaptic terminals suggested use of glutamatergic, in addition to peptidergic, neurotransmission. In summary, immunofluorescent and DiI labeling of KP neurons in thick hypothalamic sections and immunoelectron microscopic studies of KP-immunoreactive neurons in brains perfusion-fixed shortly post mortem allowed us to identify previously unexplored fine structural features of KP neurons in the mediobasal hypothalamus of humans.
Collapse
Affiliation(s)
- Szabolcs Takács
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, 43 Szigony St., Budapest, 1083, Hungary
| | - Zsuzsanna Bardóczi
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
- School of PH.D. Studies, Semmelweis University, Budapest, Hungary
| | - Katalin Skrapits
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, 43 Szigony St., Budapest, 1083, Hungary
| | - Balázs Göcz
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, 43 Szigony St., Budapest, 1083, Hungary
| | - Viktória Váczi
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, 43 Szigony St., Budapest, 1083, Hungary
| | - Zsófia Maglóczky
- Human Brain Research Laboratory, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Iván Szűcs
- Department of Pathology, St. Borbála Hospital, Tatabanya, Hungary
| | - Gergely Rácz
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - András Matolcsy
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Waljit S Dhillo
- Department of Investigative Medicine, Hammersmith Hospital, Imperial College London, London, UK
| | - Masahiko Watanabe
- Department of Anatomy, Hokkaido University School of Medicine, Sapporo, Japan
| | - Andrea Kádár
- Laboratory of Integrative Neuroendocrinology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Csaba Fekete
- Laboratory of Integrative Neuroendocrinology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Tupper Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Imre Kalló
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
- Department of Neuroscience, Faculty of Information Technology, Pázmány Péter Catholic University, Budapest, Hungary
| | - Erik Hrabovszky
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, 43 Szigony St., Budapest, 1083, Hungary.
| |
Collapse
|
39
|
Terasawa E, Garcia JP, Seminara SB, Keen KL. Role of Kisspeptin and Neurokinin B in Puberty in Female Non-Human Primates. Front Endocrinol (Lausanne) 2018; 9:148. [PMID: 29681889 PMCID: PMC5897421 DOI: 10.3389/fendo.2018.00148] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 03/20/2018] [Indexed: 11/25/2022] Open
Abstract
In human patients, loss-of-function mutations in the genes encoding kisspeptin (KISS1) and neurokinin B (NKB) and their receptors (KISS1R and NK3R, respectively) result in an abnormal timing of puberty or the absence of puberty. To understand the neuroendocrine mechanism of puberty, we investigated the contribution of kisspeptin and NKB signaling to the pubertal increase in GnRH release using rhesus monkeys as a model. Direct measurements of GnRH and kisspeptin in the median eminence of the hypothalamus with infusion of agonists and antagonists for kisspeptin and NKB reveal that kisspeptin and NKB signaling stimulate GnRH release independently or collaboratively by forming kisspeptin and NKB neuronal networks depending on the developmental age. For example, while in prepubertal females, kisspeptin and NKB signaling independently stimulate GnRH release, in pubertal females, the formation of a collaborative kisspeptin and NKB network further accelerates the pubertal increase in GnRH release. It is speculated that the collaborative mechanism between kisspeptin and NKB signaling to GnRH neurons is necessary for the complex reproductive function in females.
Collapse
Affiliation(s)
- Ei Terasawa
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI, United States
- Department of Pediatrics, University of Wisconsin, Madison, WI, United States
- *Correspondence: Ei Terasawa,
| | - James P. Garcia
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI, United States
| | - Stephanie B. Seminara
- Reproductive Endocrine Unit and the Harvard Reproductive Sciences Center, Department of Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - Kim L. Keen
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI, United States
| |
Collapse
|
40
|
Okamura H, Yamamura T, Wakabayashi Y. Mapping of KNDy neurons and immunohistochemical analysis of the interaction between KNDy and substance P neural systems in goat. J Reprod Dev 2017; 63:571-580. [PMID: 29109352 PMCID: PMC5735268 DOI: 10.1262/jrd.2017-103] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
A population of neurons in the arcuate nucleus (ARC) coexpresses kisspeptin, neurokinin B (NKB), and dynorphin, and therefore they are referred to as KNDy neurons. It has been suggested that KNDy neurons participate in several brain functions, including the control of reproduction. The present study aimed to advance our understanding of the anatomy of the KNDy neural system. We first produced an antiserum against goat kisspeptin. After confirming its specificity, the antiserum was used to histochemically detect kisspeptin-positive signals. Using the colocalization of kisspeptin and NKB immunoreactivity as a marker for KNDy neurons, we mapped distributions of their cell somata and fibers in the whole brain (except the cerebellum) of ovariectomized (OVX) goats. KNDy neuronal somata were distributed throughout the ARC, and were particularly abundant in its caudal aspect. KNDy neuronal fibers projected into several areas within the septo-preoptic-hypothalamic continuum, such as the ARC, median eminence, medial preoptic nucleus, and bed nucleus of the stria terminalis. Kisspeptin immunoreactivity was not found outside of the continuum. We then addressed to the hypothesis that substance P (SP) is also involved in the KNDy neural system. Double-labeling immunohistochemistry for kisspeptin and SP revealed that KNDy neurons did not coexpress SP, but nearly all of the KNDy neuronal somata were surrounded by fibers containing SP in the OVX goats. The present results demonstrate anatomical evidence for a robust association between the KNDy and SP neural systems.
Collapse
Affiliation(s)
- Hiroaki Okamura
- Division of Animal Breeding and Reproduction Research, Institute of Livestock and Grassland Science, NARO, Ibaraki 305-0901, Japan
| | - Takashi Yamamura
- Division of Animal Breeding and Reproduction Research, Institute of Livestock and Grassland Science, NARO, Ibaraki 305-0901, Japan
| | - Yoshihiro Wakabayashi
- Division of Animal Breeding and Reproduction Research, Institute of Livestock and Grassland Science, NARO, Ibaraki 305-0901, Japan
| |
Collapse
|
41
|
Abstract
The kisspeptin is a neuropeptide to play physiological roles in regulating gonadotropin-releasing hormone secretion in the hypothalamus. In human plasma, the kisspeptin concentration is measured, but gonadotropin-releasing hormone is not. This study aims to understand the physiological roles of the circulating kisspeptin in lactational amenorrhea in humans because prolactin reduces the kisspeptin expression and luteinizing hormone secretion resulting in anovulations in rodent brains. Plasma kisspeptin levels were measured in 11 subjects in lactational amenorrhea and in four cases with pathological amenorrhea by different etiologies for comparison using the enzyme immunoassay specific for human kisspeptin. The plasma kisspeptin levels in the 11 women with lactational amenorrhea were 15.2 ± 2.5 fmol/mL (mean ± SD) which were not significantly different as compared with 16.5 ± 4.8 fmol/mL (mean ± SD) in four age-matched women with menstrual cycles as we reported previously. In the four cases with pathological amenorrhea, their plasma kisspeptin levels were from 5.8 to 13.7 fmol/mL. This study demonstrated that the plasma kisspeptin levels were not totally reduced in lactational or pathological amenorrhea. These results suggest the physiological roles of the circulating kisspeptin are different from the role in the brain.
Collapse
Affiliation(s)
- Masato Kotani
- a Asahina Shinryoujo , Fujieda , Japan
- b Department of Internal Medicine , Fujieda Municipal General Hospital , Fujieda , Japan
- c Clinical Research Center, Shizuoka Prefectural Hospital Organization , Shizuoka , Japan
| | - Fumihiko Katagiri
- d Laboratory of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences , Hachiouji , Tokyo , Japan
| | | | - Jiro Kagawa
- f Department of Pediatrics , Fujieda Municipal General Hospital , Fujieda , Japan
| | - Issei Tanaka
- c Clinical Research Center, Shizuoka Prefectural Hospital Organization , Shizuoka , Japan
| |
Collapse
|
42
|
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: 0.6] [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]
|
43
|
Garcia JP, Guerriero KA, Keen KL, Kenealy BP, Seminara SB, Terasawa E. Kisspeptin and Neurokinin B Signaling Network Underlies the Pubertal Increase in GnRH Release in Female Rhesus Monkeys. Endocrinology 2017; 158:3269-3280. [PMID: 28977601 PMCID: PMC5659687 DOI: 10.1210/en.2017-00500] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 07/20/2017] [Indexed: 11/19/2022]
Abstract
Loss-of-function or inactivating mutations in the genes coding for kisspeptin and its receptor (KISS1R) or neurokinin B (NKB) and the NKB receptor (NK3R) in humans result in a delay in or the absence of puberty. However, precise mechanisms of kisspeptin and NKB signaling in the regulation of the pubertal increase in gonadotropin-releasing hormone (GnRH) release in primates are unknown. In this study, we conducted a series of experiments infusing agonists and antagonists of kisspeptin and NKB into the stalk-median eminence, where GnRH, kisspeptin, and NKB neuroterminal fibers are concentrated, and measuring GnRH release in prepubertal and pubertal female rhesus monkeys. Results indicate that (1) similar to those previously reported for GnRH stimulation by the KISS1R agonist (i.e., human kisspeptin-10), the NK3R agonist senktide stimulated GnRH release in a dose-responsive manner in both prepubertal and pubertal monkeys; (2) the senktide-induced GnRH release was blocked in the presence of the KISS1R antagonist peptide 234 in pubertal but not prepubertal monkeys; and (3) the kisspeptin-induced GnRH release was blocked in the presence of the NK3R antagonist SB222200 in the pubertal but not prepubertal monkeys. These results are interpreted to mean that although, in prepubertal female monkeys, kisspeptin and NKB signaling to GnRH release is independent, in pubertal female monkeys, a reciprocal signaling mechanism between kisspeptin and NKB neurons is established. We speculate that this cooperative mechanism by the kisspeptin and NKB network underlies the pubertal increase in GnRH release in female monkeys.
Collapse
Affiliation(s)
- James P Garcia
- Wisconsin National Primate Research Center, Madison, Wisconsin 53715
| | | | - Kim L Keen
- Wisconsin National Primate Research Center, Madison, Wisconsin 53715
| | - Brian P Kenealy
- Wisconsin National Primate Research Center, Madison, Wisconsin 53715
| | - Stephanie B Seminara
- Reproductive Endocrine Unit and the Harvard Reproductive Sciences Center, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114
| | - Ei Terasawa
- Wisconsin National Primate Research Center, Madison, Wisconsin 53715
- Department of Pediatrics, University of Wisconsin-Madison, Madison, Wisconsin 53706
| |
Collapse
|
44
|
Morinda Officinalis Polysaccharides Stimulate Hypothalamic GnRH Secretion in Varicocele Progression. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 2017:9057959. [PMID: 29234440 PMCID: PMC5632491 DOI: 10.1155/2017/9057959] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 08/02/2017] [Indexed: 02/07/2023]
Abstract
Varicoceles (VCs) are the predominant cause of male infertility and are a risk factor for chronic venous disease. Morinda officinalis (M. officinalis) is a traditional Chinese medicine used to tonify the kidney and strengthen yang. In this study, we evaluated the effects of water-soluble polysaccharides extracted from M. officinalis (MOPs) on gonadotropin-release hormone (GnRH) secretion in a classic experimental left VC (ELV) rat model. Intragastric administration of MOPs at a dose ranging from 50 mg kg−1 to 100 mg kg−1 facilitated improvements in sperm parameters and seminiferous epithelial structures, modulated serum hormone profiles, and stimulated GnRH synthesis and release in the hypothalamus. MOPs also promoted spinogenesis and functional spine maturation in the arcuate nuclei (Arc), wherein they acted mainly on Kiss1 and GnRH neurons. Moreover, MOP-mediated Kisspeptin-GPR54 pathway upregulation and MAPK phosphorylation activation may have been responsible for increases in GnRH synthesis and release. Collectively, the findings of this study indicate that MOPs were effective in stimulating GnRH secretion, possibly by upregulating the Kiss1/GPR54 pathway and enhancing synaptic plasticity, and that MOPs can serve as a therapy for early VCs.
Collapse
|
45
|
Rasri-Klosen K, Simonneaux V, Klosen P. Differential response patterns of kisspeptin and RFamide-related peptide to photoperiod and sex steroid feedback in the Djungarian hamster (Phodopus sungorus). J Neuroendocrinol 2017; 29. [PMID: 28834570 DOI: 10.1111/jne.12529] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 08/05/2017] [Accepted: 08/20/2017] [Indexed: 12/16/2022]
Abstract
Many animals synchronise their reproductive activity with the seasons to optimise the survival of their offspring. This synchronisation involves switching on and off their gonadotrophic axis. Ever since their discovery as key regulators of gonadotrophin-releasing hormone (GnRH) neurones, the hypothalamic RF-amide peptides kisspeptin and RFamide-related peptide (RFRP) have been a major focus of research on the seasonal regulation of the gonadotrophic axis. In the present study, we investigated the regulation of both neuropeptides in the Djungarian hamster, a major animal model for the study of seasonal reproduction. During the long-day breeding period, kisspeptin neurones in the anteroventral periventricular area are solely controlled by a positive sex steroid feedback and, in the arcuate nucleus, they are subject to a very strong negative sex steroid feedback associated with a minor photoperiodic effect. During short-day sexual quiescence, the disappearance of this hormonal feedback leads to high levels of kisspeptin in arcuate neurones. Notably, chronic central administration of kisspeptin is able to over-ride the photoperiodic inhibition of the gonadotrophic axis and reactivate the reproductive function. Therefore, our data suggest that kisspeptin secretion by arcuate neurones during sexual quiescence is inhibited by mechanisms upstream of kisspeptin neurones. RFRP expression is solely controlled by photoperiod, being strongly reduced in short days independently of the sex steroid feedback. Thus, kisspeptin and RFRP display contrasting patterns of expression and regulation. Upstream mechanisms controlling these neurones should be the focus of further studies on the roles of these RFamide neuropeptides in the seasonal control of reproduction.
Collapse
Affiliation(s)
- K Rasri-Klosen
- Institute of Cellular and Integrative Neurosciences, CNRS UPR 3212, University of Strasbourg, Strasbourg Cedex, France
- Faculty of Medicine, Department of Pre-Clinical Science, Thammasart University, Rangsit Campus, Pathumthani, Thailand
| | - V Simonneaux
- Institute of Cellular and Integrative Neurosciences, CNRS UPR 3212, University of Strasbourg, Strasbourg Cedex, France
| | - P Klosen
- Institute of Cellular and Integrative Neurosciences, CNRS UPR 3212, University of Strasbourg, Strasbourg Cedex, France
| |
Collapse
|
46
|
Aytürk N, Firat T, Kükner A, Özoğul C, Töre F, Kandirali İE, Yilmaz B. The effect of kisspeptin on spermatogenesis and apoptosis in rats. Turk J Med Sci 2017; 47:334-342. [PMID: 28263511 DOI: 10.3906/sag-1505-69] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 04/21/2016] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND/AIM To study the effect of kisspeptin, a gonadotropin release stimulator, on the testicular tissue of the rat. MATERIALS AND METHODS Four groups were formed as follows: control, Kiss-10 501397645907nmol administration for 1 day, Kiss-10 administration for 13 days, and one last group kept for 7 days following Kiss-10 applied for 13 days. Testicular tissues were stained with hematoxylin-eosin, periodic acid Schiff, Masson trichrome staining, terminal deoxynucleotidyl transferased UTP nick-end labeling, and Ki-67 immune staining. Serum testosterone levels were determined. RESULTS Serum testosterone level increased following acute application, while it was reduced by chronic treatment. Spermatogenic cells as stained by Ki-67 and TUNEL increased in the treated groups compared to the controls. Following a 7-day rest after treatment, a decrease in testosterone levels and Ki-67-stained cell numbers and an increase in TUNEL-stained cells were observed. Leydig cells showed increased vacuolization in the Kiss-1 group. Leydig cell vacuolization continued in the Kiss (13) group and was reduced in the Kiss (13 + 7) group. CONCLUSION Kiss-10 increased spermatogenic cell proliferation, while testosterone level and proliferation decreased and apoptosis increased during the waiting period.
Collapse
Affiliation(s)
- Nilüfer Aytürk
- Department of Histology and Embryology, Faculty of Medicine, Medipol University, İstanbul, Turkey
| | - Tülin Firat
- Department of Histology and Embryology, Faculty of Medicine, Abant İzzet Baysal University, Bolu, Turkey
| | - Aysel Kükner
- Department of Histology and Embryology, Faculty of Medicine, Abant İzzet Baysal University, Bolu, Turkey
| | - Candan Özoğul
- Department of Histology and Embryology, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Fatma Töre
- Department of Physiology, Faculty of Medicine, SANKO University, Gaziantep, Turkey
| | - İsmail Engin Kandirali
- Department of Urology Clinics, Bağcılar Education and Research Hospital, İstanbul, Turkey
| | - Bayram Yilmaz
- Department of Physiology, Faculty of Medicine, İstanbul Yeditepe University, İstanbul, Turkey
| |
Collapse
|
47
|
Jiang J, Jin W, Peng Y, He Z, Wei L, Li S, Wang X, Chang M, Wang R. In vivo and vitro characterization of the effects of kisspeptin-13, endogenous ligands for GPR54, on mouse gastrointestinal motility. Eur J Pharmacol 2016; 794:216-223. [PMID: 27890710 DOI: 10.1016/j.ejphar.2016.11.041] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 11/24/2016] [Accepted: 11/24/2016] [Indexed: 12/14/2022]
Abstract
Kisspeptin (KP), the endogenous ligand of GPR54, is a mammalian amidated neurohormone, which belongs to the RF-amide peptide family. However, in contrast with the related members of the RF-amide family, little information is available regarding its role in the gastrointestinal motility. With regard to the recent data suggesting KP play an important role in food intake, and while gastrointestinal motility are closely related to it. Thus, in the present work, effects of central administration of KP-13, one of the endogenous active isoforms, on gastrointestinal motility were investigated. The results indicated that intracerebroventricular (i.c.v.) infused of KP-13 significantly facilitated gastrointestinal transit, bead expulsion and fecal pellet output, respectively, while has no effect on gastric emptying. The effects were significantly reversed by GPR54 antagonist 234, but not GnRH receptor antagonist Cetrorelix. However, i.p. injected of KP-13 or compound 5 (10mg/kg), a high metabolic stability kisspeptin analog, did not affect gastrointestinal transit, suggesting that KP-13 or compound 5 facilitated gastrointestinal transit through the activation of central GPR54. Then the gastrointestinal motility-enhancing effects were also presented after infusion of KP-13 into the hypothalamus. In vitro, KP-13 (10-6M) also modulated colonic contraction, but not in the stomach and small intestine. Similarly, KP-13 (10-6M)-induced contractions of circular and longitudinal colonic muscle were significantly attenuated by antagonist 234 (10-6M). In conclusion, all the results indicated that KP-13 promoted gastrointestinal motility through the activation of GPR54, which implicate that KP/GPR54 system might be a new target to treat gastrointestinal function disorder.
Collapse
Affiliation(s)
- Jinhong Jiang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou 730000, China
| | - Weidong Jin
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou 730000, China
| | - Yali Peng
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou 730000, China
| | - Zhen He
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou 730000, China
| | - Lijuan Wei
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou 730000, China
| | - Shu Li
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou 730000, China
| | - Xiaoli Wang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou 730000, China
| | - Min Chang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou 730000, China.
| | - Rui Wang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
48
|
Cernea M, Phillips R, Padmanabhan V, Coolen LM, Lehman MN. Prenatal testosterone exposure decreases colocalization of insulin receptors in kisspeptin/neurokinin B/dynorphin and agouti-related peptide neurons of the adult ewe. Eur J Neurosci 2016; 44:2557-2568. [PMID: 27543746 PMCID: PMC5067216 DOI: 10.1111/ejn.13373] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 07/26/2016] [Accepted: 08/17/2016] [Indexed: 02/06/2023]
Abstract
Insulin serves as a link between the metabolic and reproductive systems, communicating energy availability to the hypothalamus and enabling reproductive mechanisms. Adult Suffolk ewes prenatally exposed to testosterone (T) display an array of reproductive and metabolic dysfunctions similar to those seen in women with polycystic ovarian syndrome (PCOS), including insulin resistance. Moreover, prenatal T treatment alters neuropeptide expression in KNDy (co-expressing kisspeptin, neurokinin B/dynorphin) and agouti-related peptide (AgRP) neurons in the arcuate nucleus, two populations that play key roles in the control of reproduction and metabolism, respectively. In this study, we determined whether prenatal T treatment also altered insulin receptors in KNDy and AgRP neurons, as well as in preoptic area (POA) kisspeptin, pro-opiomelanocortin (POMC), and gonadotropin-releasing hormone (GnRH) neurons of the adult sheep brain. Immunofluorescent detection of the beta subunit of insulin receptor (IRβ) revealed that KNDy, AgRP and POMC neurons, but not GnRH or POA kisspeptin neurons, colocalize IRβ in control females. Moreover, prenatal T treatment decreased the percentage of KNDy and AgRP neurons that colocalized IRβ, consistent with reduced insulin sensitivity. Administration of the anti-androgen drug, Flutamide, during prenatal T treatment, prevented the reduction in IRβ colocalization in AgRP, but not in KNDy neurons, suggesting that these effects are programmed by androgenic and oestrogenic actions, respectively. These findings provide novel insight into the effects of prenatal T treatment on hypothalamic insulin sensitivity and raise the possibility that decreased insulin receptors, specifically within KNDy and AgRP neurons, may contribute to the PCOS-like phenotype of this animal model.
Collapse
Affiliation(s)
- Maria Cernea
- Department of Neurobiology and Anatomical Sciences, The University of Mississippi Medical Center, Jackson, MS, 39232, USA
- Department of Anatomy & Cell Biology, The University of Western Ontario, London, Canada
| | - Rebecca Phillips
- Department of Neurobiology and Anatomical Sciences, The University of Mississippi Medical Center, Jackson, MS, 39232, USA
- Department of Anatomy & Cell Biology, The University of Western Ontario, London, Canada
| | - Vasantha Padmanabhan
- Department of Obstetrics and Gynecology, Pediatrics, and Reproductive Sciences Program, The University of Michigan, Ann Arbor, MI, USA
| | - Lique M Coolen
- Department of Physiology and Biophysics, The University of Mississippi Medical Center, Jackson, MS, USA
| | - Michael N Lehman
- Department of Neurobiology and Anatomical Sciences, The University of Mississippi Medical Center, Jackson, MS, 39232, USA.
| |
Collapse
|
49
|
Abstract
Groups of neurons in the hypothalamus synchronize their activity to trigger the production of hormones that sustain fertility.
Collapse
Affiliation(s)
- Sonal Shruti
- Laboratory of Development and Plasticity of the Neuroendocrine Brain and the NEUROBESE International Associated Laboratory, Institut National de la Santé et de la Recherche Médicale, Université de Lille, Unité Mixte de Recherche 1172, Lille, France
| | - Vincent Prevot
- Laboratory of Development and Plasticity of the Neuroendocrine Brain and the NEUROBESE International Associated Laboratory, Institut National de la Santé et de la Recherche Médicale, Université de Lille, Unité Mixte de Recherche 1172, Lille, France
| |
Collapse
|
50
|
Qiu J, Nestor CC, Zhang C, Padilla SL, Palmiter RD, Kelly MJ, Rønnekleiv OK. High-frequency stimulation-induced peptide release synchronizes arcuate kisspeptin neurons and excites GnRH neurons. eLife 2016; 5:e16246. [PMID: 27549338 PMCID: PMC4995096 DOI: 10.7554/elife.16246] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 07/18/2016] [Indexed: 12/19/2022] Open
Abstract
Kisspeptin (Kiss1) and neurokinin B (NKB) neurocircuits are essential for pubertal development and fertility. Kisspeptin neurons in the hypothalamic arcuate nucleus (Kiss1(ARH)) co-express Kiss1, NKB, dynorphin and glutamate and are postulated to provide an episodic, excitatory drive to gonadotropin-releasing hormone 1 (GnRH) neurons, the synaptic mechanisms of which are unknown. We characterized the cellular basis for synchronized Kiss1(ARH) neuronal activity using optogenetics, whole-cell electrophysiology, molecular pharmacology and single cell RT-PCR in mice. High-frequency photostimulation of Kiss1(ARH) neurons evoked local release of excitatory (NKB) and inhibitory (dynorphin) neuropeptides, which were found to synchronize the Kiss1(ARH) neuronal firing. The light-evoked synchronous activity caused robust excitation of GnRH neurons by a synaptic mechanism that also involved glutamatergic input to preoptic Kiss1 neurons from Kiss1(ARH) neurons. We propose that Kiss1(ARH) neurons play a dual role of driving episodic secretion of GnRH through the differential release of peptide and amino acid neurotransmitters to coordinate reproductive function.
Collapse
Affiliation(s)
- Jian Qiu
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, United States
| | - Casey C Nestor
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, United States
| | - Chunguang Zhang
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, United States
| | - Stephanie L Padilla
- Department of Biochemistry, Howard Hughes Medical Institute, University of Washington, Seattle, United States
| | - Richard D Palmiter
- Department of Biochemistry, Howard Hughes Medical Institute, University of Washington, Seattle, United States
| | - Martin J Kelly
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, United States
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, United States
| | - Oline K Rønnekleiv
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, United States
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, United States
| |
Collapse
|