1
|
Igler A, Amodei R, Roselli CE. Anatomic distribution of kisspeptin neurons in the adult sheep amygdala: Associations with sex, estrogen receptor alpha, androgen receptor, and sexual partner preference. J Neuroendocrinol 2025; 37:e70011. [PMID: 40033683 PMCID: PMC12045730 DOI: 10.1111/jne.70011] [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: 09/18/2024] [Revised: 02/07/2025] [Accepted: 02/16/2025] [Indexed: 03/05/2025]
Abstract
Kisspeptin neurons are primarily known for regulating reproductive function by stimulating hormone release that controls puberty and fertility. While typically associated with the hypothalamus, recent research suggests their presence in other brain regions, including the amygdala. The amygdala, crucial for emotional processing and social behaviors, consists of various nuclei. However, the specific distribution and potential functional implications of kisspeptin neurons within this region remain unclear. Understanding kisspeptin neuron distribution in the sheep amygdala could provide insights into their roles in modulating reproductive functions, emotional, and social behaviors in a species closely related to humans. This study employed immunohistochemistry and RNAscope™ fluorescent in situ hybridization to map the distribution of kisspeptin fibers and cells in the amygdala of intact adult male and luteal-phase female sheep. The research also investigated the co-expression of Kiss1 with estrogen receptor-α (ESR1) and androgen receptor (AR) mRNA, as well as the presence of kisspeptin receptor (Kiss1r) mRNA-containing cells. Kisspeptin immunoreactive fibers were most dense in the medial amygdala, while Kiss1 mRNA-containing cells were abundant in the medial, cortical, and basal nuclei. Extensive co-expression of Kiss1 with ESR1 and AR mRNA was observed. In the posterior medial nucleus, 80% of kisspeptin neurons co-expressed ESR1, and 40% co-expressed AR. Kiss1r mRNA-containing cells were found in the medial, cortical, and basal nuclei and co-localized within cells expressing Kiss1 mRNA. No differences in kisspeptin cell numbers were found between rams and ewes or between rams with different sexual partner preferences. This study provides a foundational map of the kisspeptin system in the sheep amygdala, offering insights into its potential roles in reproductive, emotional, and social behaviors. The extensive co-expression of Kiss1 mRNA with ESR1 and AR mRNA suggests possible regulation by sex steroids, while the presence of Kiss1r mRNA-containing cells indicates potential autocrine or paracrine signaling. These findings contribute to our understanding of kisspeptin neurons' distribution and potential functions beyond the hypothalamus, particularly in the amygdala.
Collapse
Affiliation(s)
- Anna Igler
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon, USA
| | - Rebecka Amodei
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon, USA
| | - Charles E Roselli
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
2
|
Koysombat K, Tsoutsouki J, Patel AH, Comninos AN, Dhillo WS, Abbara A. Kisspeptin and neurokinin B: roles in reproductive health. Physiol Rev 2025; 105:707-764. [PMID: 39813600 DOI: 10.1152/physrev.00015.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 10/17/2024] [Accepted: 11/13/2024] [Indexed: 01/18/2025] Open
Abstract
Kisspeptin and neurokinin B (NKB) play a key role in several physiological processes including in puberty, adult reproductive function including the menstrual cycle, as well as mediating the symptoms of menopause. Infundibular kisspeptin neurons, which coexpress NKB, regulate the activity of gonadotropin-releasing hormone (GnRH) neurons and thus the physiological pulsatile secretion of GnRH from the hypothalamus. Outside of their hypothalamic reproductive roles, these peptides are implicated in several physiological functions including sexual behavior and attraction, placental function, and bone health. Over the last two decades, research findings have considerably enhanced our understanding of the physiological regulation of the hypothalamic-pituitary-gonadal (HPG) axis and identified potential therapeutic applications. For example, recognition of the role of kisspeptin as the natural inductor of ovulation has led to research investigating its use as a safer, more physiological trigger of oocyte maturation in in vitro fertilization (IVF) treatment. Moreover, the key role of NKB in the pathophysiology of menopausal hot flashes has led to the development of pharmacological antagonism of this pathway. Indeed, fezolinetant, a neurokinin 3 receptor antagonist, has recently received Food and Drug Administration (FDA) approval for clinical use to treat menopausal vasomotor symptoms. Here, we discuss the roles of kisspeptin and NKB in human physiology, including in the regulation of puberty, menstrual cyclicity, reproductive behavior, pregnancy, menopause, and bone homeostasis. We describe how perturbations of these key physiological processes can result in disease states and consider how kisspeptin and NKB could be exploited diagnostically as well as therapeutically to treat reproductive disorders.
Collapse
Affiliation(s)
- Kanyada Koysombat
- Department of Investigative Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Jovanna Tsoutsouki
- Department of Investigative Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Aaran H Patel
- Department of Investigative Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Alexander N Comninos
- Department of Investigative Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Waljit S Dhillo
- Department of Investigative Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Ali Abbara
- Department of Investigative Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, United Kingdom
| |
Collapse
|
3
|
Watanabe Y, Fisher L, Campbell RE, Jasoni CL. Developmental expression patterns of gonadal hormone receptors in arcuate kisspeptin and GABA neurons of the postnatal female mouse. J Neuroendocrinol 2025; 37:e13477. [PMID: 39605295 DOI: 10.1111/jne.13477] [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: 07/18/2024] [Revised: 10/29/2024] [Accepted: 11/17/2024] [Indexed: 11/29/2024]
Abstract
The arcuate nucleus of the hypothalamus (ARC) is central in the neuronal regulation of fertility and reproduction through translating gonadal steroid hormone cues into the GnRH signaling pathway in the brain. Evidence suggests that circulating gonadal steroids play an important role in modulating female reproduction via kisspeptin and γ-aminobutyric acid (GABA) neurons in the ARC in both development and adulthood. However, the temporal onset of these ARC neurons' sensitivity to gonadal steroids is unknown. Using RNAscope® in situ hybridization, we localized androgen receptor (Ar), estrogen receptor alpha (Esr1), and progesterone receptor (Pgr) expression in ARC kisspeptin or GABA neurons of female mice at postnatal day (P)4, P8, P12, P20, and P60. A probe that binds to kiss1 mRNA or vGat mRNA was used to produce signal in kisspeptin or GABA neurons, respectively. In adult, we identified that the vast majority of kisspeptin neurons coexpressed Esr1 (95%) and Pgr (93%), while a smaller proportion coexpressed Ar (66%). Similar proportions of Ar- or Esr1-positive kisspeptin neurons were seen from P4, suggesting that kisspeptin neurons develop adult-like sensitivity to androgen and estrogen in early postnatal life. In contrast, the proportion of Pgr-positive kisspeptin cells in early life was significantly lower than in adulthood, suggesting that progesterone sensitivity develops over time in the ARC kisspeptin population. ARC GABA neurons also colocalized with Ar (70%), Esr1 (64%), or Pgr (85%) in adulthood. GABA neurons continuously expressed Esr1 or Pgr from the postnatal stages to adulthood, while the proportion of Ar-positive GABA neurons gradually increased from P4 (24%) to P20 (59%). These results suggest that while ARC GABA neurons can respond to circulating estrogen and progesterone from early postnatal ages, this same population may become more sensitive to androgens during later postnatal life. Our findings identified the expression patterns of Ar, Esr1, and Pgr by ARC kisspeptin and GABA neurons during early postnatal life. These data provide the understanding for the hormone sensitivity of these populations during early postnatal life, the critical time for the formation and regulation of female reproductive physiology.Esr1 (95%) and Pgr (93%), while a smaller proportion coexpressed Ar (66%). Similar proportions of Ar- or Esr1-positive kisspeptin neurons were seen from P4, suggesting that kisspeptin neurons develop adult-like sensitivity to androgen and estrogen in early postnatal life. In contrast, the proportion of Pgr-positive kisspeptin cells in early life was significantly lower than in adulthood, suggesting that progesterone sensitivity develops over time in the ARC kisspeptin population. ARC GABA neurons also colocalized with Ar (70%), Esr1 (64%), or Pgr (85%) in adulthood. GABA neurons continuously expressed Esr1 or Pgr from the postnatal stages to adulthood, while the proportion of Ar-positive GABA neurons gradually increased from P4 (24%) to P20 (59%). These results suggest that while ARC GABA neurons can respond to circulating estrogen and progesterone from early postnatal ages, this same population may become more sensitive to androgens during later postnatal life. Our findings identified the expression patterns of Ar, Esr1, and Pgr by ARC kisspeptin and GABA neurons during early postnatal life. These data provide the understanding for the hormone sensitivity of these populations during early postnatal life, the critical time for the formation and regulation of female reproductive physiology.
Collapse
Affiliation(s)
- Yugo Watanabe
- Centre for Neuroendocrinology, Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
- Department of Biochemistry, School of Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Lorryn Fisher
- Centre for Neuroendocrinology, Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Rebecca E Campbell
- Centre for Neuroendocrinology, Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Christine L Jasoni
- Centre for Neuroendocrinology, Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, New Zealand
| |
Collapse
|
4
|
González-Flores O, Domínguez-Ordóñez R, Delgado-Macuil RJ, Tlachi-López JL, Luna-Hernández A, Montes-Narváez O, Pfaus JG, García-Juárez M. Participation of kisspeptin, progesterone, and GnRH receptors on lordosis behavior induced by kisspeptin. Physiol Behav 2024; 283:114609. [PMID: 38851441 DOI: 10.1016/j.physbeh.2024.114609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/27/2024] [Accepted: 06/05/2024] [Indexed: 06/10/2024]
Abstract
The neuropeptide kisspeptin (Kiss) is crucial in regulating the hypothalamic-pituitary-gonadal axis. It is produced by two main groups of neurons in the hypothalamus: the rostral periventricular region around the third ventricle and the arcuate nucleus. Kiss is the peptide product of the KiSS-1 gene and serves as the endogenous agonist for the GPR54 receptor. The Kiss/GPR54 system functions as a critical regulator of the reproductive system. Thus, we examined the effect of intracerebroventricular administration of 3 μg of Kiss to the right lateral ventricle of ovariectomized rats primed with a dose of 5 μg subcutaneous (sc) of estradiol benzoate (EB). Kiss treatment increased the lordosis quotient at all times tested. However, the lordosis reflex score was comparatively lower yet still significant compared to the control group. To investigate receptor specificity and downstream mechanisms on lordosis, we infused 10 μg of GPR54 receptor antagonist, Kiss-234, 5 μg of the progestin receptor antagonist, RU486, or 3 μg of antide, a gonadotropin-releasing hormone-1 (GnRH-1) receptor antagonist, to the right lateral ventricle 30 min before an infusion of 3 μg of Kiss. Results demonstrated a significant reduction in the facilitation of lordosis behavior by Kiss at 60 and 120 min when Kiss-234, RU486, or antide were administered. These findings suggest that Kiss stimulates lordosis expression by activating GPR54 receptors on GnRH neurons and that Kiss/GPR54 system is an essential intermediary by which progesterone activates GnRH.
Collapse
Affiliation(s)
- Oscar González-Flores
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, Tlaxcala, México
| | - Raymundo Domínguez-Ordóñez
- Licenciatura en Ingeniería Agronómica y Zootecnia, CRC, Benemérita Universidad Autónoma de, Puebla, México
| | - Raul Jacobo Delgado-Macuil
- Centro de Investigación en Biotecnología Aplicada, Instituto Politécnico Nacional, Santa Inés, Tecuexcomac, Tlaxcala, México
| | | | - Ailyn Luna-Hernández
- Doctorado en Ciencias Biológicas, Universidad Autónoma de Tlaxcala, Tlaxcala, México
| | - Omar Montes-Narváez
- Doctorado en Ciencias Biológicas, Universidad Autónoma de Tlaxcala, Tlaxcala, México
| | - James G Pfaus
- Center for Sexual Health and Intervention, Czech National Institute of Mental Health, Klecany, Czech Republic; Department of Psychology and Life Sciences, Charles University, Prague, Czech Republic
| | - Marcos García-Juárez
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, Tlaxcala, México.
| |
Collapse
|
5
|
Dillon KM, Lohr DB, Novak AG, Petriv AMV, Neifert NT, Moore AM. Deletion of Nuclear Progesterone Receptors From Kisspeptin Cells Does Not Impair Negative Feedback in Female Mice. Endocrinology 2024; 165:bqae121. [PMID: 39253941 DOI: 10.1210/endocr/bqae121] [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: 07/10/2024] [Revised: 09/03/2024] [Accepted: 09/08/2024] [Indexed: 09/11/2024]
Abstract
Reproductive function in mammals depends on the ability of progesterone (P4) to suppress pulsatile gonadotrophin-releasing hormone (GnRH) and luteinizing hormone (LH) secretion in a homeostatic-negative feedback loop. Previous research identified that cells upstream from GnRH neurons expressing the nuclear progesterone receptor (PGR) are required for P4-negative feedback. However, the identity of these cells and the mechanism by which they reduce GnRH/LH pulsatile secretion is unknown. We aimed to address the hypothesis that PGR expressed by a neural population in the arcuate nucleus recently identified as the GnRH pulse generator, cells expressing kisspeptin, neurokinin B, and dynorphin (KNDy cells), mediate P4-negative feedback. To achieve this, we used female mice with the PGR gene conditionally deleted from kisspeptin cells (KPRKO mice) and observed a substantial decrease in the percentage of KNDy neurons coexpressing PGR messenger RNA (mRNA) (11% in KPRKO mice vs 86% in wild-type [WT] mice). However, KPRKO mice did not display changes in the frequency or amplitude of LH pulses in diestrus or estrus, nor in the ability of exogenous P4 to blunt a postcastration increase in LH. Further, mRNA expression of arcuate kisspeptin and dynorphin, which are excitatory and inhibitory to GnRH secretion, respectively, remained unaltered in KPRKO mice compared to WT controls. Together, these findings show that the near-complete loss of PGR signaling from KNDy cells does not affect negative feedback regulation of GnRH pulse generation in mice, suggesting that feedback through this receptor can occur via a small number of KNDy cells or a yet unidentified cell population.
Collapse
Affiliation(s)
- Kendra M Dillon
- Department of Biological Sciences, Brain Health Research Institute, Kent State University, Kent, OH 44242-0001, USA
| | - Dayanara B Lohr
- Department of Biological Sciences, Brain Health Research Institute, Kent State University, Kent, OH 44242-0001, USA
| | - Alyssa G Novak
- Department of Biological Sciences, Brain Health Research Institute, Kent State University, Kent, OH 44242-0001, USA
| | - Anna-Maria V Petriv
- Department of Biological Sciences, Brain Health Research Institute, Kent State University, Kent, OH 44242-0001, USA
| | - Nicole T Neifert
- Department of Biological Sciences, Brain Health Research Institute, Kent State University, Kent, OH 44242-0001, USA
| | - Aleisha M Moore
- Department of Biological Sciences, Brain Health Research Institute, Kent State University, Kent, OH 44242-0001, USA
| |
Collapse
|
6
|
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
|
7
|
Wu X, Zhang Z, Li Y, Zhao Y, Ren Y, Tian Y, Hou M, Guo Y, Li Q, Tian W, Jiang R, Zhang Y, Gong Y, Li H, Li G, Liu X, Kang X, Li D, Tian Y. Estrogen promotes gonadotropin-releasing hormone expression by regulating tachykinin 3 and prodynorphin systems in chicken. Poult Sci 2024; 103:103820. [PMID: 38759565 PMCID: PMC11127269 DOI: 10.1016/j.psj.2024.103820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/12/2024] [Accepted: 05/02/2024] [Indexed: 05/19/2024] Open
Abstract
The "KNDy neurons" located in the hypothalamic arcuate nucleus (ARC) of mammals are known to co-express kisspeptin, neurokinin B (NKB), and dynorphin (DYN), and have been identified as key mediators of the feedback regulation of steroid hormones on gonadotropin-releasing hormone (GnRH). However, in birds, the genes encoding kisspeptin and its receptor GPR54 are genomic lost, leaving unclear mechanisms for feedback regulation of GnRH by steroid hormones. Here, the genes tachykinin 3 (TAC3) and prodynorphin (PDYN) encoding chicken NKB and DYN neuropeptides were successfully cloned. Temporal expression profiling indicated that TAC3, PDYN and their receptor genes (TACR3, OPRK1) were mainly expressed in the hypothalamus, with significantly higher expression at 30W than at 15W. Furthermore, overexpression or interference of TAC3 and PDYN can regulate the GnRH mRNA expression. In addition, in vivo and in vitro assays showed that estrogen (E2) could promote the mRNA expression of TAC3, PDYN, and GnRH, as well as the secretion of GnRH/LH. Mechanistically, E2 could dimerize the nuclear estrogen receptor 1 (ESR1) to regulate the expression of TAC3 and PDYN, which promoted the mRNA and protein expression of GnRH gene as well as the secretion of GnRH. In conclusion, these results revealed that E2 could regulate the GnRH expression through TAC3 and PDYN systems, providing novel insights for reproductive regulation in chickens.
Collapse
Affiliation(s)
- Xing Wu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Zihao Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Yijie Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Yudian Zhao
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Yangguang Ren
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Yixiang Tian
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang 453003, China
| | - Meng Hou
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Yulong Guo
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Qi Li
- Henan zhumadian agricultural school, zhumadian, 463000, China
| | - Weihua Tian
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Ruirui Jiang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Yanhua Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Yujie Gong
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Hong Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Guoxi Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Xiaojun Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Xiangtao Kang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Donghua Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Yadong Tian
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China.
| |
Collapse
|
8
|
Condi FLDF, Fuchs LFP, Carvalho KC, Baracat EC. Treatment with Raloxifene Induces the Expression of Kisspeptin, Insulin, and Androgen Receptors in Bones of Castrated Adult Female Rats. Rev Bras Ortop 2024; 59:e228-e234. [PMID: 38606141 PMCID: PMC11006519 DOI: 10.1055/s-0044-1779319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 06/26/2023] [Indexed: 04/13/2024] Open
Abstract
Objective To evaluate the effects of estrogen, raloxifene and genistein on the expression of KISS1 (kisspeptin), KISS1R (kisspeptin receptor), AR (androgen receptor) and INSR (insulin receptor) in the bones of ovariectomized rats. Methods Forty-eight adult rats were randomly divided into 6 groups, containing 8 animals each: G1-nonovariectomized control; G2-ovariectomized and treated with conjugated equine estrogens (50 µg/Kg/day); G3-ovariectomized and treated with raloxifene (0.75 mg/kg/day); G4-ovariectomized animal that received soy extract with genistein (300 mg/kg/day); G5-ovariectomized animal that received estrogen and genistein; and G6-ovariectomized animal that received estrogen and raloxifene. Three months after surgery, the castrated animals received the drugs orally daily for 120 days. All animals were sacrificed after this period, by deepening the anesthesia. The left tibia was removed for total RNA extraction and analysis of gene expression of KISS1 , KISS1R , AR and INSR , by quantitative real-time polymerase chain reaction (qRT-PCR). Results KISS1 was not detected in any of the treated groups. KISS1R , INSR and AR showed higher expression in the G3 group ( p < 0.001), while lower levels of transcripts for these genes were observed in G4 and G5. G2 animals showed hypoexpression of the evaluated genes. Conclusion The results indicate that raloxifene, alone or combined with estrogen, was able to induce the expression of genes associated with the recovery of bone tissue homeostasis in ovariectomized rats.
Collapse
Affiliation(s)
| | - Luiz Fernando Portugal Fuchs
- Departamento de Ginecologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - Katia Candido Carvalho
- Laboratório de Ginecologia Estrutural e Molecular (LIM 58), Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brasil
| | - Edmund Chada Baracat
- Laboratório de Ginecologia Estrutural e Molecular (LIM 58), Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brasil
| |
Collapse
|
9
|
Yacca SS, Kanasaki H, Tumurbaatar T, Cairang Z, Oride A, Okada H, Kyo S. Changes in pituitary gonadotropin subunits and hypothalamic Kiss-1 gene expression by administration of sex steroids in ovary-intact female rats. Endocrine 2024; 83:733-746. [PMID: 37966704 DOI: 10.1007/s12020-023-03596-0] [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: 05/28/2023] [Accepted: 11/03/2023] [Indexed: 11/16/2023]
Abstract
OBJECTIVE We examined how the sex steroids influence the synthesis of gonadotropins. MATERIALS AND METHODS The effects of sex steroids estradiol (E2), progesterone (P4), and dihydrotestosterone (DHT) in pituitary gonadotroph cell model (LβT2 cells) in vitro and ovary-intact rats in vivo were examined. The effects of sex steroids on Kiss1 gene expression in the hypothalamus were also examined in ovary-intact rats. RESULTS In LβT2 cells, E2 increased common glycoprotein alpha (Cga) and luteinizing hormone beta (Lhb) subunit promoter activity as well as their mRNA expression. Although gonadotropin subunit promoter activity was not modulated by P4, Cga and Lhb mRNA expression was increased by P4. DHT inhibited Cga and Lhb mRNA expression with a concomitant decrease in their promoter activity. During the 2-week administration of exogenous E2 to ovary-intact rats, the estrous cycle determined by vaginal smears was disrupted. P4 or DHT administration completely eliminated the estrous cycle. Protein expression of all three gonadotropin subunits within the pituitary gland was inhibited by E2 or P4 treatment in vivo; however, DHT reduced Cga expression but did not modulate Lhb or follicle-stimulating hormone beta subunit expression. E2 administration significantly repressed Kiss1 mRNA expression in a posterior hypothalamic region that included the arcuate nucleus. P4 and DHT did not modulate Kiss1 mRNA expression in this region. In contrast, P4 administration significantly inhibited Kiss1 mRNA expression in the anterior region of the hypothalamus that included the anteroventral periventricular nucleus. The expression of gonadotropin-releasing hormone (Gnrh) mRNA in the anterior hypothalamic region, where the preoptic area is located, appeared to be decreased by treatment with E2 and P4. CONCLUSION Our findings suggest that sex steroids have different effects in the hypothalamus and pituitary gland.
Collapse
Affiliation(s)
- Susdiaman S Yacca
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, 693-8501, Japan
| | - Haruhiko Kanasaki
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, 693-8501, Japan.
| | - Tuvshintugs Tumurbaatar
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, 693-8501, Japan
| | - Zhuoma Cairang
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, 693-8501, Japan
| | - Aki Oride
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, 693-8501, Japan
| | - Hiroe Okada
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, 693-8501, Japan
| | - Satoru Kyo
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, 693-8501, Japan
| |
Collapse
|
10
|
Voigt C, Gahr M, Bennett NC. Differential regulation of Kiss1 gene expression by oestradiol in the hypothalamus of the female Damaraland mole-rat, an induced ovulator. Gen Comp Endocrinol 2023; 341:114334. [PMID: 37302764 DOI: 10.1016/j.ygcen.2023.114334] [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: 02/08/2023] [Revised: 04/25/2023] [Accepted: 06/06/2023] [Indexed: 06/13/2023]
Abstract
Kisspeptin, a product of the Kiss1 gene is considered a potent stimulator of gonadotropin release, by interacting with its receptor, the G protein-coupled receptor 54. Kiss1 neurons are known to mediate the positive and negative feedback effects of oestradiol on GnRH neurons that control the pulsatile and surge secretion of GnRH. While in spontaneously ovulating mammals the GnRH/LH surge is initiated by a rise in ovarian oestradiol secreted from maturing follicles, in induced ovulators, the primary trigger is the mating stimulus. Damaraland mole rats (Fukomys damarensis) are cooperatively breeding, subterranean rodents that exhibit induced ovulation. We have previously described in this species the distribution and differential expression pattern of Kiss1-expressing neurons in the hypothalamus of males and females. Here we examine whether oestradiol (E2) regulates the hypothalamic Kiss1 expression in a similar way as described for spontaneously ovulating rodent species. By means of in situ hybridisation, we measured Kiss1 mRNA among groups of ovary-intact, ovariectomized (OVX) and OVX females treated with E2 (OVX + E2). In the arcuate nucleus (ARC), Kiss1 expression increased after ovariectomy and decreased with E2 treatment. In the preoptic region, Kiss1 expression after gonadectomy was similar to the level of wild-caught gonad-intact controls, but was dramatically upregulated with E2 treatment. The data suggest that, similar to other species, Kiss1 neurons in the ARC, which are inhibited by E2, play a role in the negative feedback control on GnRH release. The exact role of the Kiss1 neuron population in the preoptic region, which is stimulated by E2, remains to be determined.
Collapse
Affiliation(s)
- Cornelia Voigt
- Department of Zoology and Entomology, University of Pretoria, 0028 Pretoria, South Africa.
| | - Manfred Gahr
- Department of Behavioural Neurobiology, Max Planck Institute for Biological Intelligence, D-82319 Seewiesen, Germany.
| | - Nigel C Bennett
- Department of Zoology and Entomology, University of Pretoria, 0028 Pretoria, South Africa.
| |
Collapse
|
11
|
Meunier MA, Porte C, Poissenot K, Vacher H, Brachet M, Chamero P, Beltramo M, Abecia JA, Delgadillo JA, Chemineau P, Keller M. Male-induced early puberty correlates with the maturation of arcuate nucleus kisspeptin neurons in does. J Neuroendocrinol 2023; 35:e13284. [PMID: 37157154 DOI: 10.1111/jne.13284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 03/09/2023] [Accepted: 04/17/2023] [Indexed: 05/10/2023]
Abstract
In goats, early exposure of spring-born females to sexually active bucks induces an early puberty onset assessed by the first ovulation. This effect is found when females are continuously exposed well before the male breeding season starting in September. The first aim of this study was to evaluate whether a shortened exposure of females to males could also lead to early puberty. We assessed the onset of puberty in Alpine does isolated from bucks (ISOL), exposed to wethers (CAS), exposed to intact bucks from the end of June (INT1), or mid-August (INT2). Intact bucks became sexually active in mid-September. At the beginning of October, 100% of INT1 and 90% of INT2 exposed does ovulated, in contrast to the ISOL (0%) and CAS (20%) groups. This demonstrated that contact with males that become sexually active is the main factor prompting precocious puberty in females. Furthermore, a reduced male exposure during a short window before the breeding season is sufficient to induce this phenomenon. The second aim was to investigate the neuroendocrine changes induced by male exposure. We found a significant increase in kisspeptin immunoreactivity (fiber density and number of cell bodies) in the caudal part of the arcuate nucleus of INT1 and INT2 exposed females. Thus, our results suggest that sensory stimuli from sexually active bucks (e.g., chemosignals) may trigger an early maturation of the ARC kisspeptin neuronal network leading to gonadotropin-releasing hormone secretion and first ovulation.
Collapse
Affiliation(s)
- Maxime A Meunier
- UMR Physiologie de la Reproduction et des Comportements, INRAE, CNRS, IFCE, Université de Tours, Nouzilly, France
| | - Chantal Porte
- UMR Physiologie de la Reproduction et des Comportements, INRAE, CNRS, IFCE, Université de Tours, Nouzilly, France
| | - Kévin Poissenot
- UMR Physiologie de la Reproduction et des Comportements, INRAE, CNRS, IFCE, Université de Tours, Nouzilly, France
| | - Hélène Vacher
- UMR Physiologie de la Reproduction et des Comportements, INRAE, CNRS, IFCE, Université de Tours, Nouzilly, France
| | - Morgane Brachet
- UMR Physiologie de la Reproduction et des Comportements, INRAE, CNRS, IFCE, Université de Tours, Nouzilly, France
| | - Pablo Chamero
- UMR Physiologie de la Reproduction et des Comportements, INRAE, CNRS, IFCE, Université de Tours, Nouzilly, France
| | - Massimiliano Beltramo
- UMR Physiologie de la Reproduction et des Comportements, INRAE, CNRS, IFCE, Université de Tours, Nouzilly, France
| | - José A Abecia
- Departamento de Producción Animal y Ciencia de los Alimentos, IUCA, Universidad de Zaragoza, Zaragoza, Spain
| | - José A Delgadillo
- Centro de Investigación en Reproducción Caprina, Universidad Autónoma Agraria Antonio Narro, Torreón, Mexico
| | - Philippe Chemineau
- UMR Physiologie de la Reproduction et des Comportements, INRAE, CNRS, IFCE, Université de Tours, Nouzilly, France
| | - Matthieu Keller
- UMR Physiologie de la Reproduction et des Comportements, INRAE, CNRS, IFCE, Université de Tours, Nouzilly, France
| |
Collapse
|
12
|
Prashar V, Arora T, Singh R, Sharma A, Parkash J. Hypothalamic Kisspeptin Neurons: Integral Elements of the GnRH System. Reprod Sci 2023; 30:802-822. [PMID: 35799018 DOI: 10.1007/s43032-022-01027-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/23/2022] [Indexed: 12/19/2022]
Abstract
Highly sophisticated and synchronized interactions of various cells and hormonal signals are required to make organisms competent for reproduction. GnRH neurons act as a common pathway for multiple cues for the onset of puberty and attaining reproductive function. GnRH is not directly receptive to most of the signals required for the GnRH secretion during the various phases of the ovarian cycle. Kisspeptin neurons of the hypothalamus convey these signals required for the synchronized release of the GnRH. The steroid-sensitive anteroventral periventricular nucleus (AVPV) kisspeptin and arcuate nucleus (ARC) KNDy neurons convey steroid feedback during the reproductive cycle necessary for GnRH surge and pulse, respectively. AVPV region kisspeptin neurons also communicate with nNOS synthesizing neurons and suprachiasmatic nucleus (SCN) neurons to coordinate the process of the ovarian cycle. Neurokinin B (NKB) and dynorphin play roles in the GnRH pulse stimulation and inhibition, respectively. The loss of NKB and kisspeptin function results in the development of neuroendocrine disorders such as hypogonadotropic hypogonadism (HH) and infertility. Ca2+ signaling is essential for GnRH pulse generation, which is propagated through gap junctions between astrocytes-KNDy and KNDy-KNDy neurons. Impaired functioning of KNDy neurons could develop the characteristics associated with polycystic ovarian syndrome (PCOS) in rodents. Kisspeptin-increased synthesis led to excessive secretion of the LH associated with PCOS. This review provides the latest insights and understanding into the role of the KNDy and AVPV/POA kisspeptin neurons in GnRH secretion and PCOS.
Collapse
Affiliation(s)
- Vikash Prashar
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Tania Arora
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Randeep Singh
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Arti Sharma
- Department of Computational Sciences, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Jyoti Parkash
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India.
| |
Collapse
|
13
|
Hellier V, Dardente H, Lomet D, Cognié J, Dufourny L. Interactions between β-endorphin and kisspeptin neurons of the ewe arcuate nucleus are modulated by photoperiod. J Neuroendocrinol 2023; 35:e13242. [PMID: 36880357 DOI: 10.1111/jne.13242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023]
Abstract
Opioid peptides are well-known modulators of the central control of reproduction. Among them, dynorphin coexpressed in kisspeptin (KP) neurons of the arcuate nucleus (ARC) has been thoroughly studied for its autocrine effect on KP release through κ opioid receptors. Other studies have suggested a role for β-endorphin (BEND), a peptide cleaved from the pro-opiomelanocortin precursor, on food intake and central control of reproduction. Similar to KP, BEND content in the ARC of sheep is modulated by day length and BEND modulates food intake in a dose-dependent manner. Because KP levels in the ARC vary with photoperiodic and metabolic status, a photoperiod-driven influence of BEND neurons on neighboring KP neurons is plausible. The present study aimed to investigate a possible modulatory action of BEND on KP neurons located in the ovine ARC. Using confocal microscopy, numerous KP appositions on BEND neurons were found but there was no photoperiodic variation of the number of these interactions in ovariectomized, estradiol-replaced ewes. By contrast, BEND terminals on KP neurons were twice as numerous under short days, in ewes having an activated gonadotropic axis, compared to anestrus ewes under long days. Injection of 5 μg BEND into the third ventricle of short-day ewes induced a significant and specific increase of activated KP neurons (16% vs. 9% in controls), whereas the percentage of overall activated (c-Fos positive) neurons, was similar between both groups. These data suggest a photoperiod-dependent influence of BEND on KP neurons of the ARC, which may influence gonadotropin-releasing hormone pulsatile secretion and inform KP neurons about metabolic status.
Collapse
Affiliation(s)
- Vincent Hellier
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France
| | - Hugues Dardente
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France
| | - Didier Lomet
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France
| | - Juliette Cognié
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France
| | | |
Collapse
|
14
|
Epigenetic Regulation of miR-25 and Lnc107153 on Expression of Seasonal Estrus Key Gene CHGA in Sheep. BIOLOGY 2023; 12:biology12020250. [PMID: 36829527 PMCID: PMC9952879 DOI: 10.3390/biology12020250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/29/2023] [Accepted: 02/02/2023] [Indexed: 02/09/2023]
Abstract
Pituitary pars tuberalis (PT) plays an important role as the transmission center in the seasonal reproduction of animals. It helps convert external photoperiod signals into intrinsic seasonal reproduction signals. In sheep PT, specific expression patterns of several genes (including short photoperiod-induced gene CHGA and long photoperiod genes EYA3 and TSHβ) under different photoperiods are crucial characteristics during this signal transduction. Recent studies have revealed the role of epigenetics in regulating the expression of seasonal reproductive key genes. Therefore, we explored whether microRNAs and LncRNAs regulated the expressions of the above key genes. Firstly, the expression of miR-25 and CHGA showed a significant negative correlation in sheep PT. Results of the dual luciferase reporter assay and miR-25 overexpression indicated that miR-25 could inhibit the expression of CHGA by specifically binding to its 3'UTR region in pituitary cells. Then, expression negative correlation and dual luciferase reporter analyses were used to screen and identify the candidate LncRNA (Lnc107153) targeted by miR-25. Finally, the results of fluorescence in situ hybridization and Lnc107153 overexpression suggested that Lnc107153 and miR-25 were involved in the epigenetic regulation of CHGA expression. However, the expressions of EYA3 and TSHβ were not regulated by miRNAs. These results will provide new insights into the epigenetic regulatory network of key genes in sheep seasonal reproduction.
Collapse
|
15
|
McQuillan HJ, Clarkson J, Kauff A, Han SY, Yip SH, Cheong I, Porteous R, Heather AK, Herbison AE. Definition of the estrogen negative feedback pathway controlling the GnRH pulse generator in female mice. Nat Commun 2022; 13:7433. [PMID: 36460649 PMCID: PMC9718805 DOI: 10.1038/s41467-022-35243-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 11/23/2022] [Indexed: 12/03/2022] Open
Abstract
The mechanisms underlying the homeostatic estrogen negative feedback pathway central to mammalian fertility have remained unresolved. Direct measurement of gonadotropin-releasing hormone (GnRH) pulse generator activity in freely behaving mice with GCaMP photometry demonstrated striking estradiol-dependent plasticity in the frequency, duration, amplitude, and profile of pulse generator synchronization events. Mice with Cre-dependent deletion of ESR1 from all kisspeptin neurons exhibited pulse generator activity identical to that of ovariectomized wild-type mice. An in vivo CRISPR-Cas9 approach was used to knockdown ESR1 expression selectively in arcuate nucleus (ARN) kisspeptin neurons. Mice with >80% deletion of ESR1 in ARN kisspeptin neurons exhibited the ovariectomized pattern of GnRH pulse generator activity and high frequency LH pulses but with very low amplitude due to reduced responsiveness of the pituitary. Together, these studies demonstrate that estrogen utilizes ESR1 in ARN kisspeptin neurons to achieve estrogen negative feedback of the GnRH pulse generator in mice.
Collapse
Affiliation(s)
- H James McQuillan
- Centre for Neuroendocrinology, University of Otago School of Biomedical Sciences, Dunedin, 9054, New Zealand.,Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, 9054, New Zealand
| | - Jenny Clarkson
- Centre for Neuroendocrinology, University of Otago School of Biomedical Sciences, Dunedin, 9054, New Zealand.,Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, 9054, New Zealand
| | - Alexia Kauff
- Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, 9054, New Zealand
| | - Su Young Han
- Centre for Neuroendocrinology, University of Otago School of Biomedical Sciences, Dunedin, 9054, New Zealand.,Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, 9054, New Zealand.,Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EG, UK
| | - Siew Hoong Yip
- Centre for Neuroendocrinology, University of Otago School of Biomedical Sciences, Dunedin, 9054, New Zealand.,Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, 9054, New Zealand
| | - Isaiah Cheong
- Centre for Neuroendocrinology, University of Otago School of Biomedical Sciences, Dunedin, 9054, New Zealand.,Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, 9054, New Zealand
| | - Robert Porteous
- Centre for Neuroendocrinology, University of Otago School of Biomedical Sciences, Dunedin, 9054, New Zealand.,Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, 9054, New Zealand
| | - Alison K Heather
- Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, 9054, New Zealand
| | - Allan E Herbison
- Centre for Neuroendocrinology, University of Otago School of Biomedical Sciences, Dunedin, 9054, New Zealand. .,Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, 9054, New Zealand. .,Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EG, UK.
| |
Collapse
|
16
|
Exposure of rams in sexual rest to sexually activated males in spring increases plasma LH and testosterone concentrations. Theriogenology 2022; 192:116-121. [PMID: 36088808 DOI: 10.1016/j.theriogenology.2022.08.035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/23/2022]
Abstract
Eight stimulating rams, and twelve stimulated rams, were used to determine whether a similar endocrine response to the introduction of sexually active males in spring in a flock of ewes is observed in a flock of rams. The stimulating rams (n = 4) were induced into a sexually active state by exposure to 2 months of long days (16 h light/d) (15 December-15 February). At the end of the long-day period, rams were returned to the natural photoperiod. Control-stimulating rams (n = 4) were kept under the natural photoperiod. On April 20, stimulated rams were divided into 2 groups, and joined with activated (ACT; n = 6) or control stimulating rams (C; n = 6). On the day of ram introduction, stimulated rams were blood sampled for 8 h at 20-min intervals, from 4 h before to 4 h after ram introduction, and next day from 24 to 28 h after ram introduction, and analyzed for plasma LH concentrations, and 10, 20 and 30 days after ram introduction to measure plasma testosterone levels. Mean (±SEM) plasma LH concentrations (ng/ml) of stimulated rams were similar during the 4 h before stimulating-ram introduction (ACT: 0.59 ± 0.03; C: 0.53 ± 0.04; P > 0.05). The introduction of the photoperiod-treated stimulating rams increased LH concentrations of stimulated rams during the 4 h after their introduction (1.14 ± 0.37) compared with the C group (0.51 ± 0.03; P < 0.05), especially during the first hour (ACT: 0.93 ± 0.16; C: 0.49 ± 0.03; P < 0.05), and during the blood sampling period 24-28 h after ram introduction (0.75 ± 0.07 vs. 0.58 ± 0.04; P < 0.05). Before the introduction of stimulating rams, the LH pulse frequencies and amplitudes did not differ between groups; however, LH pulsatility was higher at 4 h (0.58 ± 0.11 pulses/h; P < 0.05), and had trend to be higher 24 h (0.50 ± 0.06) (P = 0.10) after the introduction of the photoperiod-treated stimulating rams compared with the control-stimulating rams (0.29 ± 0.08 and 0.29 ± 0.10, respectively). As for LH pulses, there was an effect of group (P < 0.05) on LH amplitude, which presented a trend to be higher in ACT rams 4 h after ram introduction (1.68 ± 0.30; P < 0.10) and higher 24 h (1.07 ± 0.08; P < 0.05) after ram introduction, compared with LH amplitudes of C rams (0.71 ± 0.06 and 0.82 ± 0.07, respectively). Plasma testosterone concentrations of rams exposed to photoperiod-treated activated rams were higher than those of rams exposed to control-stimulating rams, at 4 h, 20 and 30 days after ram introduction (P < 0.05). In conclusion, sexually active rams in spring are able to stimulate LH and testosterone secretion of other rams in sexual rest, a phenomenon we called "ram-to-ram effect".
Collapse
|
17
|
Delgadillo JA, Espinoza-Flores LA, Abecia JA, Hernández H, Keller M, Chemineau P. Sexually active male goats stimulate the endocrine and sexual activities of other males in seasonal sexual rest through the "buck-to-buck effect". Domest Anim Endocrinol 2022; 81:106746. [PMID: 35750019 DOI: 10.1016/j.domaniend.2022.106746] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/30/2022] [Accepted: 05/31/2022] [Indexed: 11/28/2022]
Abstract
Male goats rendered sexually active by exposure to a photoperiodic treatment are more efficient than untreated goats in stimulating LH secretion and ovulation in seasonally anestrous goats. This phenomenon is called the "male effect." Here, we determined whether sexually active bucks are able to stimulate the endocrine and sexual activities of other bucks in seasonal sexual rest through the phenomenon that we called the "buck-to-buck effect." We used bucks rendered sexually active (SA) during sexual rest by exposure to 2.5 mo of artificial long days (16 h of light per d) and untreated, sexually inactive (SI) bucks. In Experiment 1, we determined the short-term (21 d) LH and testosterone responses of sexually inactive bucks joined with a SA or SI buck. In Experiment 2, we determined the long-term (60 d) testosterone and sexual behavior responses of sexually inactive bucks joined with 2 SA or SI bucks. In Experiment 3, we determined the efficacy of bucks initially exposed to the buck-to-buck effect, the SABB bucks, to thereafter induce a "classical" male effect in seasonally anestrous goats. In Experiments 1 and 2, there was an interaction between time and groups in LH and testosterone plasma concentrations (P < 0.01). In Experiment 1, plasma LH concentrations were greater in bucks joined with a SA buck than in those joined with an SI buck (P < 0.05). In Experiments 1 and 2, testosterone concentrations were greater in bucks joined with SA bucks than in those joined with SI bucks (P < 0.05). In addition, in Experiment 2, the sexually inactive bucks joined with SA bucks displayed more nudging than those joined with SI bucks (P < 0.001). In Experiment 3, kidding rates did not differ between females joined with SA (34 of 40: 85%) or SABB bucks (32 of 40: 80%; P > 0.05). We concluded that the endocrine and sexual activities of bucks during sexual rest can be stimulated by SA bucks. In addition, SABB bucks are able to stimulate the reproductive activity of seasonally anestrous goats.
Collapse
Affiliation(s)
- J A Delgadillo
- Centro de Investigación en Reproducción Caprina (CIRCA), Universidad Autónoma Agraria Antonio Narro, Torreón, Coahuila, Mexico.
| | - L A Espinoza-Flores
- Centro de Investigación en Reproducción Caprina (CIRCA), Universidad Autónoma Agraria Antonio Narro, Torreón, Coahuila, Mexico
| | - J A Abecia
- Departamento de Producción Animal y Ciencia de los Alimentos, IUCA, Universidad de Zaragoza, Zaragoza, Spain
| | - H Hernández
- Centro de Investigación en Reproducción Caprina (CIRCA), Universidad Autónoma Agraria Antonio Narro, Torreón, Coahuila, Mexico
| | - M Keller
- UMR Physiologie de la Reproduction et des Comportements, INRAE, CNRS, IFCE, Université de Tours, Agreenium, 37380 Nouzilly, France
| | - P Chemineau
- UMR Physiologie de la Reproduction et des Comportements, INRAE, CNRS, IFCE, Université de Tours, Agreenium, 37380 Nouzilly, France
| |
Collapse
|
18
|
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: 19] [Impact Index Per Article: 6.3] [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
|
19
|
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
|
20
|
Zubair H, Shamas S, Ullah H, Nabi G, Huma T, Ullah R, Hussain R, Shahab M. Morphometric and Myelin Basic Protein Expression Changes in Arcuate Nucleus Kisspeptin Neurons Underlie Activation of Hypothalamic Pituitary Gonadal-axis in Monkeys ( Macaca Mulatta) during the Breeding Season. Endocr Res 2022; 47:113-123. [PMID: 35866239 DOI: 10.1080/07435800.2022.2102649] [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] [Indexed: 11/03/2022]
Abstract
INTRODUCTION Kisspeptin is involved in the hypothalamic pituitary gonadal-axis' seasonal regulation in rodents and sheep. Studies of kisspeptin signaling in regulating the transition between breeding and nonbreeding seasons have focused on kisspeptin expression, myelin basic protein (MBP) expression around kisspeptin-ir cells, and quantifying the synaptic connections between kisspeptin and gonadotropin-releasing hormone (GnRH) neurons in various animal models; however, the role of kisspeptin in regulating the seasonal breeding of primates has not been explored yet. OBJECTIVE This study investigated changes in kisspeptin signaling during breeding and a non-breeding season in a non-human primate model, the rhesus monkey. METHODS Three adult male monkeys (n = 3) from the breeding season and two monkeys (n = 2) from the non-breeding season were used in this study. After measuring the testicular volume and collecting a single blood sample, all animals were humanely euthanized under controlled conditions, and their hypothalami were collected and processed. Two 20 µm thick hypothalamic sections (mediobasal hypothalamus) from each animal were processed for kisspeptin-MBP and kisspeptin-GnRH immunohistochemistry (IHC). One section from each animal was used as a primary antibody omitted control to check the nonspecific binding in each IHC. RESULTS Compared to the non-breeding season, plasma testosterone levels and testicular volumes were significantly higher in monkeys during the breeding season. Furthermore, compared to the non-breeding season, increased kisspeptin expression and a higher number of synaptic contacts between kisspeptin fibers and GnRH cell bodies were observed in the arcuate nucleus of the breeding season monkeys. In contrast, enlarged kisspeptin soma and higher MBP expression were observed in non-breeding monkeys. CONCLUSION Our results indicated enhanced kisspeptin signaling in primate hypothalamus during the breeding season. These findings support the idea that kisspeptin acts as a mediator for the seasonal regulation of the reproductive axis in higher primates.
Collapse
Affiliation(s)
- Hira Zubair
- Laboratory of Reproductive Neuroendocrinology, Department of Zoology, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Shazia Shamas
- Laboratory of Reproductive Neuroendocrinology, Department of Zoology, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
- Department of Zoology, University of Gujrat, Hafiz Hayat Campus, Gujrat, Pakistan
| | - Hamid Ullah
- Laboratory of Reproductive Neuroendocrinology, Department of Zoology, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Ghulam Nabi
- Institute of Nature Conservation, Polish Academy of Sciences, Krakow, Poland
| | - Tanzeel Huma
- Laboratory of Reproductive Neuroendocrinology, Department of Zoology, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Rahim Ullah
- Laboratory of Reproductive Neuroendocrinology, Department of Zoology, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
- Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, ZJ, China
| | - Rashad Hussain
- Department of Neurology, Center for Translational Neuro-medicine, University of Rochester, Rochester, NY, USA
| | - Muhammad Shahab
- Laboratory of Reproductive Neuroendocrinology, Department of Zoology, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| |
Collapse
|
21
|
Pal P, Aggarwal A, Deb R. Effects of photoperiod on reproduction of cattle: a review. BIOL RHYTHM RES 2022. [DOI: 10.1080/09291016.2022.2102707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Prasanna Pal
- Animal Physiology Division, ICAR-National Dairy Research Institute, Karnal, India
| | - Anjali Aggarwal
- Animal Physiology Division, ICAR-National Dairy Research Institute, Karnal, India
| | - Rajib Deb
- Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, India
| |
Collapse
|
22
|
Sheep as a model for neuroendocrinology research. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 189:1-34. [PMID: 35595346 DOI: 10.1016/bs.pmbts.2022.01.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Animal models remain essential to understand the fundamental mechanisms of physiology and pathology. Particularly, the complex and dynamic nature of neuroendocrine cells of the hypothalamus make them difficult to study. The neuroendocrine systems of the hypothalamus are critical for survival and reproduction, and are highly conserved throughout vertebrate evolution. Their roles in controlling body metabolism, growth and body composition, stress, electrolyte balance, and reproduction, have been intensively studied, and have yielded groundbreaking discoveries. Many of these discoveries would not have been feasible without the use of the domestic sheep (Ovis aries). The sheep has been used for decades to study the neuroendocrine systems of the hypothalamus and has become a model for human neuroendocrinology. The aim of this chapter is to review some of the profound biomedical discoveries made possible by the use of sheep. The advantages and limitations of sheep as a neuroendocrine model will be discussed. While no animal model can perfectly recapitulate a human disease or condition, sheep are invaluable for enabling manipulations not possible in human subjects and isolating physiologic variables to garner insight into neuroendocrinology and associated pathologies.
Collapse
|
23
|
Dardente H, Simonneaux V. GnRH and the photoperiodic control of seasonal reproduction: Delegating the task to kisspeptin and RFRP-3. J Neuroendocrinol 2022; 34:e13124. [PMID: 35384117 DOI: 10.1111/jne.13124] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/22/2022] [Accepted: 03/03/2022] [Indexed: 10/18/2022]
Abstract
Synchronization of mammalian breeding activity to the annual change of photoperiod and environmental conditions is of the utmost importance for individual survival and species perpetuation. Subsequent to the early 1960s, when the central role of melatonin in this adaptive process was demonstrated, our comprehension of the mechanisms through which light regulates gonadal activity has increased considerably. The current model for the photoperiodic neuroendocrine system points to pivotal roles for the melatonin-sensitive pars tuberalis (PT) and its seasonally-regulated production of thyroid-stimulating hormone (TSH), as well as for TSH-sensitive hypothalamic tanycytes, radial glia-like cells located in the basal part of the third ventricle. Tanycytes respond to TSH through increased expression of thyroid hormone (TH) deiodinase 2 (Dio2), which leads to heightened production of intrahypothalamic triiodothyronine (T3) during longer days of spring and summer. There is strong evidence that this local, long-day driven, increase in T3 links melatonin input at the PT to gonadotropin-releasing hormone (GnRH) output, to align breeding with the seasons. The mechanism(s) through which T3 impinges upon GnRH remain(s) unclear. However, two distinct neuronal populations of the medio-basal hypothalamus, which express the (Arg)(Phe)-amide peptides kisspeptin and RFamide-related peptide-3, appear to be well-positioned to relay this seasonal T3 message towards GnRH neurons. Here, we summarize our current understanding of the cellular, molecular and neuroendocrine players, which keep track of photoperiod and ultimately govern GnRH output and seasonal breeding.
Collapse
Affiliation(s)
- Hugues Dardente
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France
| | - Valérie Simonneaux
- Institute for Cellular and Integrative Neuroscience, University of Strasbourg, Strasbourg, France
| |
Collapse
|
24
|
He X, Di R, Guo X, Cao X, Zhou M, Li X, Xia Q, Wang X, Zhang J, Zhang X, Liu Q, Chu M. Transcriptomic Changes of Photoperiodic Response in the Hypothalamus Were Identified in Ovariectomized and Estradiol-Treated Sheep. Front Mol Biosci 2022; 9:848144. [PMID: 35480892 PMCID: PMC9036065 DOI: 10.3389/fmolb.2022.848144] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 02/28/2022] [Indexed: 01/11/2023] Open
Abstract
Accurate timing of seasonal changes is an essential ability for an animal’s survival, and the change in the photoperiod is the key factor affecting reproductive seasonality in mammals. Emerging evidence has suggested that multiple hypothalamic genes participate in the photoperiod-induced regulation of reproductive activities in sheep, but the mechanism is still unclear. In this study, we initially examined the plasma level of two major reproductive hormones, namely, follicle-stimulating hormone (FSH) and prolactin (PRL), under different photoperiods in ovariectomized and estradiol-treated (OVX + E2) sheep using radioimmunoassay (RIA). Of the two hormones, the concentration of PRL significantly increased with the extension of the photoperiod, while FSH showed the opposite trend. Subsequently, an examination of the transcriptomic variation between the short photoperiod (SP) and long photoperiod (LP) was conducted. Differential expression analyses and functional annotation showed that several key genes in the insulin secretion (VAMP2, PRKACB, PRKCG, and PLCB1), GnRH (MAPK13, CGA, CDC42, ATF4, and LHB) pathways, and circadian entrainment (KCNJ5, PER1, GNB2, MTNR1A, and RASD1), as well as numerous lncRNAs, including XR_173257.3, XR_173415.3, XR_001435315.1, XR_001024596.2, and XR_001023464.2, were shown potentially vital for the hypothalamic photoperiodic response. Four of the differentially expressed mRNAs and lncRNAs were validated by qPCR. The constructed mRNA–mRNA interaction networks further revealed that transcripts potentially participated in hypothalamic thyroid hormone synthesis, endocrine resistance, and neuroactive ligand–receptor interactions. The interactome analysis of lncRNAs and their targets implied that XR_173257.3 and its target arylalkylamine N-acetyltransferase (AANAT) and XR_173415.3 and its target TH might participate in the regulation of seasonal reproduction. Together, the changes in reproductive hormones and transcriptome will help to determine the important photoperiod-induced lncRNAs and mRNAs and provide a valuable resource for further research on reproductive seasonality in sheep.
Collapse
Affiliation(s)
- Xiaoyun He
- Key Laboratory of Animal Genetics and Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Ran Di
- Key Laboratory of Animal Genetics and Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xiaofei Guo
- Institute of Animal Husbandry and Veterinary Medicine, Tianjin Academy of Agricultural Sciences, Tianjin, China
| | - Xiaohan Cao
- Key Laboratory of Animal Genetics and Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Mei Zhou
- Key Laboratory of Animal Genetics and Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xiaoyu Li
- Key Laboratory of Animal Genetics and Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Qing Xia
- Key Laboratory of Animal Genetics and Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xiangyu Wang
- Key Laboratory of Animal Genetics and Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jinlong Zhang
- Institute of Animal Husbandry and Veterinary Medicine, Tianjin Academy of Agricultural Sciences, Tianjin, China
| | - Xiaosheng Zhang
- Institute of Animal Husbandry and Veterinary Medicine, Tianjin Academy of Agricultural Sciences, Tianjin, China
| | - Qiuyue Liu
- Key Laboratory of Animal Genetics and Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- *Correspondence: Qiuyue Liu, ; Mingxing Chu,
| | - Mingxing Chu
- Key Laboratory of Animal Genetics and Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- *Correspondence: Qiuyue Liu, ; Mingxing Chu,
| |
Collapse
|
25
|
Grassi D, Marraudino M, Garcia-Segura LM, Panzica GC. The hypothalamic paraventricular nucleus as a central hub for the estrogenic modulation of neuroendocrine function and behavior. Front Neuroendocrinol 2022; 65:100974. [PMID: 34995643 DOI: 10.1016/j.yfrne.2021.100974] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 12/18/2021] [Accepted: 12/21/2021] [Indexed: 12/17/2022]
Abstract
Estradiol and hypothalamic paraventricular nucleus (PVN) help coordinate reproduction with body physiology, growth and metabolism. PVN integrates hormonal and neural signals originating in the periphery, generating an output mediated both by its long-distance neuronal projections, and by a variety of neurohormones produced by its magnocellular and parvocellular neurosecretory cells. Here we review the cyto-and chemo-architecture, the connectivity and function of PVN and the sex-specific regulation exerted by estradiol on PVN neurons and on the expression of neurotransmitters, neuromodulators, neuropeptides and neurohormones in PVN. Classical and non-classical estrogen receptors (ERs) are expressed in neuronal afferents to PVN and in specific PVN interneurons, projecting neurons, neurosecretory neurons and glial cells that are involved in the input-output integration and coordination of neurohormonal signals. Indeed, PVN ERs are known to modulate body homeostatic processes such as autonomic functions, stress response, reproduction, and metabolic control. Finally, the functional implications of the estrogenic modulation of the PVN for body homeostasis are discussed.
Collapse
Affiliation(s)
- D Grassi
- Department of Anatomy, Histology and Neuroscience, Universidad Autonoma de Madrid, Madrid, Spain
| | - M Marraudino
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Torino, Italy
| | - L M Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - G C Panzica
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Torino, Italy; Department of Neuroscience Rita Levi Montalcini, University of Torino, Torino, Italy.
| |
Collapse
|
26
|
Clarke IJ, Reed CB, Burke CR, Li Q, Meier S. Kiss1 expression in the hypothalamic arcuate nucleus is lower in dairy cows of reduced fertility. Biol Reprod 2022; 106:802-813. [PMID: 34982141 PMCID: PMC9040656 DOI: 10.1093/biolre/ioab240] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/13/2021] [Accepted: 12/21/2021] [Indexed: 11/13/2022] Open
Abstract
We tested the hypothesis that divergent genetic merit for fertility of dairy cows is due to aberrant reproductive neuroendocrine function. The kisspeptin status of non-pregnant cows of either positive (POS) or negative (NEG) breeding values (BVs) for fertility was studied in three groups (n = 8), based on their previous post-partum period: POS cows, which had spontaneous ovarian cycles (POS-CYC) and NEG cows, which either cycled (NEG-CYC) or did not cycle (NEG-NONCYC). Ovarian cycles were synchronized, blood samples were taken to define endocrine status, and the animals were slaughtered in an artificial follicular phase. The brains and the pituitary glands were collected for quantitative polymerase chain reaction (qPCR) and in situ hybridization of hypothalamic GNRH1, Kiss1, TAC3, and PDYN and pituitary expression of LHB and FSHB. Gonadotropin releasing hormone (GnRH) and kisspeptin levels were quantified in snap frozen median eminence (ME). GNRH1 expression and GnRH levels in the ME were similar across groups. Kiss1 expression in the preoptic area of the hypothalamus was also similar across groups, but Kiss1 in the arcuate nucleus was almost 2-fold higher in POS-CYC cows than in NEG groups. TAC3 expression was higher in POS-CYC cows. The number of pituitary gonadotropes and the level of expression of LHB and FSHB were similar across groups. We conclude that the lower levels of Kiss1 and TAC3 in NEG cows with low fertility status and may lead to deficient GnRH and gonadotropin secretion.
Collapse
Affiliation(s)
- Iain J Clarke
- Neuroscience Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, Victoria, Australia, 3800
| | | | - Chris R Burke
- DairyNZ Limited, Private Bag 3221, Hamilton 3240, New Zealand
| | - Qun Li
- Neuroscience Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, Victoria, Australia, 3800
| | - Susanne Meier
- DairyNZ Limited, Private Bag 3221, Hamilton 3240, New Zealand
| |
Collapse
|
27
|
Khan S, Batool B, Zubair H, Bano R, Ahmad S, Shahab M. Expression and co-localization of RFRP-3 and kisspeptin during breeding and non-breeding season in the hypothalamus of male rhesus monkey ( Macaca mulatta). Reprod Med Biol 2022; 21:e12479. [PMID: 35847413 PMCID: PMC9270642 DOI: 10.1002/rmb2.12479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/12/2022] [Accepted: 06/21/2022] [Indexed: 12/01/2022] Open
Abstract
Propose The mechanism that underpins how RFRP-3 and kisspeptin interacts are not fully understood in higher primates. This study therefore set out to assess RFRP-3 and kisspeptin expression and their morphological interactions in the breeding, and in the non-breeding period in monkey hypothalamus. Methods Eight mature male macaques (Macaca mulatta) in the breeding season (February; n = 4) and non-breeding season (June; n = 4) were used. To reveal the expression and co-localization of RFRP-3 and kisspeptin, double-labeled immunohistochemistry was performed. Testicular volume, sperm count, and plasma testosterone level were also measured to validate the breeding and non-breeding paradigms. Results Testicular volume, plasma testosterone level, and sperm count showed a significant reduction during non-breeding season. The number of kisspeptin-positive cells was significantly increased during the breeding season (p < 0.05), whereas more RFRP-3-positive cell bodies were seen in the non-breeding season (p < 0.01). Close contacts of RFRP-3 fibers with kisspeptin cells showed no significant difference (p > 0.05) across seasons. However, co-localization of RFRP-3-ir cell bodies onto kisspeptin IR cell bodies showed a statistical increase (p < 0.01) in non-breeding season. Conclusion In higher primates, RFRP-3 decreases kisspeptin drives from the same cells to GnRH neurons in an autocrine manner causing suppression of the reproductive axis during the non-breeding period.
Collapse
Affiliation(s)
- Safdar Khan
- Department of Zoology, Laboratory of Reproductive Neuroendocrinology, Faculty of Biological SciencesQuaid‐i‐Azam UniversityIslamabadPakistan
| | - Bakhtwar Batool
- Department of Zoology, Laboratory of Reproductive Neuroendocrinology, Faculty of Biological SciencesQuaid‐i‐Azam UniversityIslamabadPakistan
| | - Hira Zubair
- Department of Zoology, Laboratory of Reproductive Neuroendocrinology, Faculty of Biological SciencesQuaid‐i‐Azam UniversityIslamabadPakistan
| | - Riffat Bano
- Department of Zoology, Laboratory of Reproductive Neuroendocrinology, Faculty of Biological SciencesQuaid‐i‐Azam UniversityIslamabadPakistan
| | - Shakil Ahmad
- Department of Zoology, Laboratory of Reproductive Neuroendocrinology, Faculty of Biological SciencesQuaid‐i‐Azam UniversityIslamabadPakistan
| | - Muhammad Shahab
- Department of Zoology, Laboratory of Reproductive Neuroendocrinology, Faculty of Biological SciencesQuaid‐i‐Azam UniversityIslamabadPakistan
- Shaheed Benazir Bhutto University, SheringalDir UpperPakistan
| |
Collapse
|
28
|
Ozawa H. Kisspeptin neurons as an integration center of reproductive regulation: Observation of reproductive function based on a new concept of reproductive regulatory nervous system. Reprod Med Biol 2021; 21:e12419. [PMID: 34934400 PMCID: PMC8656200 DOI: 10.1002/rmb2.12419] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/20/2021] [Accepted: 09/24/2021] [Indexed: 11/29/2022] Open
Abstract
Background Regulation of the reproductive system has been explained by the actions and feedback of gonadotropin releasing hormone‐luteinizing hormone/follicle stimulating hormone (GnRH‐LH/FSH) ‐sex steroids; however, the discovery of kisspeptin neurons and a kisspeptin‐GnRH‐LH/FSH axis has prompted this regulation to be reviewed. Methods We investigated changes in kisspeptin neurons and associated changes in the hypothalamic‐pituitary‐gonadal (HPG) axis under various situations and experimental conditions using histochemical methods. Main findings (Results) Kisspeptin neurons play an important role in receiving and integrating information from internal and external environmental factors and communicating it to the conventional HPG axis. Conclusion The recently described Kisspeptin‐GnRH‐LH/FSH‐gonad system regulates reproductive function via mechanisms that until recently were not completely understood.
Collapse
Affiliation(s)
- Hitoshi Ozawa
- Department of Anatomy and Neurobiology Graduate School of Medicine Nippon Medical School Tokyo Japan
| |
Collapse
|
29
|
Moore AM, Lohr DB, Coolen LM, Lehman MN. Prenatal Androgen Exposure Alters KNDy Neurons and Their Afferent Network in a Model of Polycystic Ovarian Syndrome. Endocrinology 2021; 162:bqab158. [PMID: 34346492 PMCID: PMC8402932 DOI: 10.1210/endocr/bqab158] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Indexed: 02/08/2023]
Abstract
Polycystic ovarian syndrome (PCOS), the most common endocrinopathy affecting women worldwide, is characterized by elevated luteinizing hormone (LH) pulse frequency due to the impaired suppression of gonadotrophin-releasing hormone (GnRH) release by steroid hormone negative feedback. Although neurons that co-express kisspeptin, neurokinin B, and dynorphin (KNDy cells) were recently defined as the GnRH/LH pulse generator, little is understood about their role in the pathogenesis of PCOS. We used a prenatal androgen-treated (PNA) mouse model of PCOS to determine whether changes in KNDy neurons or their afferent network underlie altered negative feedback. First, we identified elevated androgen receptor gene expression in KNDy cells of PNA mice, whereas progesterone receptor and dynorphin gene expression was significantly reduced, suggesting elevated androgens in PCOS disrupt progesterone negative feedback via direct actions upon KNDy cells. Second, we discovered GABAergic and glutamatergic synaptic input to KNDy neurons was reduced in PNA mice. Retrograde monosynaptic tract-tracing revealed a dramatic reduction in input originates from sexually dimorphic afferents in the preoptic area, anteroventral periventricular nucleus, anterior hypothalamic area and lateral hypothalamus. These results reveal 2 sites of neuronal alterations potentially responsible for defects in negative feedback in PCOS: changes in gene expression within KNDy neurons, and changes in synaptic inputs from steroid hormone-responsive hypothalamic regions. How each of these changes contribute to the neuroendocrine phenotype seen in in PCOS, and the role of specific sets of upstream KNDy afferents in the process, remains to be determined.
Collapse
Affiliation(s)
- Aleisha M Moore
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
- Brain Health Research Institute, Kent State University, Kent, OH 44242, USA
| | - Dayanara B Lohr
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
- Brain Health Research Institute, Kent State University, Kent, OH 44242, USA
| | - Lique M Coolen
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
- Brain Health Research Institute, Kent State University, Kent, OH 44242, USA
| | - Michael N Lehman
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
- Brain Health Research Institute, Kent State University, Kent, OH 44242, USA
| |
Collapse
|
30
|
Yang H, Fu L, Luo Q, Li L, Zheng F, Liu X, Zhao Z, Wang Z, Xu H. Comparative Analysis and Identification of Differentially Expressed microRNAs in the Hypothalamus of Kazakh Sheep Exposed to Different Photoperiod Conditions. BIOCHEMISTRY. BIOKHIMIIA 2021; 86:1315-1325. [PMID: 34903161 DOI: 10.1134/s0006297921100126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
MicroRNAs (miRNA) plays an important role in several mammalian biological regulatory processes by post-transcriptionally regulating gene expression. However, there is little information on the miRNAs involved in the photoperiodism pathway that controls seasonal activity. To enhance our knowledge on the effect of different photoperiod conditions on miRNA, we divided Kazakh sheep into two groups: one exposed to a long photoperiod (LP, 16L:8D) and another with exposed to a short photoperiod (SP, 8L:16D) under supplemental feeding conditions. Further we compared the related miRNAs and target genes between the two groups. Fifteen differentially expressed miRNAs were identified, which were associated with 310 regulatory pathways covering photoperiodism, reproductive hormones, and nutrition. The miR-136-GNAQ pair was selected and validated as a differentially expressed, and a dual-luciferase reporter assay showed that the negative feedback loop existed between them. Examination of the expression profile revealed that the GNAQ expression was low in the estrous females both under LP and SP conditions, but high expression of GNAQ was observed in the anestrous females under LP conditions. Moreover, functional analysis revealed that KISS1 and GnRH expression was upregulated when GNAQ expression was downregulated in the hypothalamic cells, whereas DIO2 and TSHB expression was downregulated. Thus, miR-136-GNAQ might act as a switch in the regulation of seasonal estrus under different photoperiod conditions. These findings further enrich our understanding of the relationship between miRNAs and seasonal regulation of reproductive activity. Furthermore, our study provides novel insights into the miRNA-mediated regulatory mechanisms for overcoming photoinhibition in the seasonally breeding mammals, such as Kazakh sheep.
Collapse
Affiliation(s)
- Heng Yang
- College of Veterinary Medicine, Southwest University, Chongqing, 404100, China. .,Immunology Research Center, Medical Research Institute, Southwest University, Chongqing, 404100, China
| | - Lin Fu
- Research Institute of Herbivorous Livestock, Chongqing Academy of Animal Sciences, Chongqing, 404100, China
| | - Qifeng Luo
- College of Veterinary Medicine, Southwest University, Chongqing, 404100, China
| | - Licai Li
- College of Veterinary Medicine, Southwest University, Chongqing, 404100, China
| | - Fangling Zheng
- College of Veterinary Medicine, Southwest University, Chongqing, 404100, China
| | - Xianxia Liu
- College of Animal Science and Technology, Shihezi University, Xinjiang, 830000, China
| | - Zongsheng Zhao
- College of Animal Science and Technology, Shihezi University, Xinjiang, 830000, China
| | - Zhiying Wang
- College of Veterinary Medicine, Southwest University, Chongqing, 404100, China
| | - Huihao Xu
- College of Veterinary Medicine, Southwest University, Chongqing, 404100, China.
| |
Collapse
|
31
|
Gloria A, Contri A, Mele E, Fasano S, Pierantoni R, Meccariello R. Kisspeptin Receptor on the Sperm Surface Reflects Epididymal Maturation in the Dog. Int J Mol Sci 2021; 22:ijms221810120. [PMID: 34576283 PMCID: PMC8466692 DOI: 10.3390/ijms221810120] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 09/14/2021] [Accepted: 09/16/2021] [Indexed: 11/16/2022] Open
Abstract
Alongside the well-known central modulatory role, the Kisspeptin system, comprising Kiss1, its cleavage products (Kisspeptins), and Kisspeptin receptor (Kiss1R), was found to regulate gonadal functions in vertebrates; however, its functional role in the male gamete and its localization during maturation have been poorly understood. The present study analyzed Kisspeptin system in dog testis and spermatozoa recovered from different segments of the epididymis, with focus on Kiss1R on sperm surface alongside the maturation during epididymal transit, demonstrated by modification in sperm kinetic, morphology, and protamination. The proteins Kiss1 and Kiss1R were detected in dog testis. The receptor Kiss1R only was detected in total protein extracts from epididymis spermatozoa, whereas dot blot revealed Kiss1 immunoreactivity in the epidydimal fluid. An increase of the Kiss1R protein on sperm surface along the length of the epididymis, with spermatozoa in the tail showing plasma membrane integrity and Kiss1R protein (p < 0.05 vs. epididymis head and body) was observed by flow cytometry and further confirmed by epifluorescence microscopy and Western blot carried on sperm membrane preparations. In parallel, during the transit in the epididymis spermatozoa significantly modified their ability to move and the pattern of motility; a progressive increase in protaminization also occurred. In conclusion, Kisspeptin system was detected in dog testis and spermatozoa. Kiss1R trafficking toward plasma membrane along the length of the epididymis and Kiss1 in epididymal fluid suggested a new functional role of the Kisspeptin system in sperm maturation and storage.
Collapse
Affiliation(s)
- Alessia Gloria
- Faculty of Veterinary Medicine, University of Teramo, Loc. Piano d’Accio, 64100 Teramo, Italy;
| | - Alberto Contri
- Faculty of Biosciences and Technologies for Agriculture Food and Environment, University of Teramo, Via Balzarini 1, 64100 Teramo, Italy
- Correspondence: (A.C.); (R.M.)
| | - Elena Mele
- Department of Movement Sciences and Wellbeing, Parthenope University of Naples, 80133 Naples, Italy;
| | - Silvia Fasano
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (S.F.); (R.P.)
| | - Riccardo Pierantoni
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (S.F.); (R.P.)
| | - Rosaria Meccariello
- Department of Movement Sciences and Wellbeing, Parthenope University of Naples, 80133 Naples, Italy;
- Correspondence: (A.C.); (R.M.)
| |
Collapse
|
32
|
Prashar V, Arora T, Singh R, Sharma A, Parkash J. Interplay of KNDy and nNOS neurons: A new possible mechanism of GnRH secretion in the adult brain. Reprod Biol 2021; 21:100558. [PMID: 34509713 DOI: 10.1016/j.repbio.2021.100558] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 08/25/2021] [Accepted: 08/28/2021] [Indexed: 02/07/2023]
Abstract
Reproduction in mammals is favoured when there is sufficient energy available to permit the survival of offspring. Neuronal nitric oxide synthase expressing neurons produce nitric oxide in the proximity of the gonadotropin-releasing hormone neurons in the preoptic region. nNOS neurons are an integral part of the neuronal network controlling ovarian cyclicity and ovulation. Nitric oxide can directly regulate the activity of the GnRH neurons and play a vital role neuroendocrine axis. Kisspeptin neurons are essential for the GnRH pulse and surge generation. The anteroventral periventricular nucleus (AVPV), kisspeptin neurons are essential for GnRH surge generation. KNDy neurons are present in the hypothalamus's arcuate nucleus (ARC), co-express NKB and dynorphin, essential for GnRH pulse generation. Kisspeptin-neurokinin B-dynorphin (KNDy) neuroendocrine molecules of the hypothalamus are key components in the central control of GnRH secretion. The hypothalamic neurons kisspeptin, KNDy, nitric oxide synthase (NOS), and other mediators such as leptin, adiponectin, and ghrelin, play an active role in attaining puberty. Kisspeptin signalling is mediated by NOS, which further results in the secretion of GnRH. Neuronal nitric oxide is critical for attaining puberty, but its direct role in adult GnRH secretion is poorly understood. This review mainly focuses on the role of nNOS and its interplay with KNDy neurons in the hormonal regulation of reproduction.
Collapse
Affiliation(s)
- Vikash Prashar
- Department of Zoology, School of Basic and Applied Sciences, Central University Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Tania Arora
- Department of Zoology, School of Basic and Applied Sciences, Central University Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Randeep Singh
- Department of Zoology, School of Basic and Applied Sciences, Central University Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Arti Sharma
- Department of Computational Biology, School of Basic and Applied Sciences, Central University Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Jyoti Parkash
- Department of Zoology, School of Basic and Applied Sciences, Central University Punjab, Ghudda, Bathinda, 151401, Punjab, India.
| |
Collapse
|
33
|
Hernández-Hernández JM, Martin GB, Becerril-Pérez CM, Pro-Martínez A, Cortez-Romero C, Gallegos-Sánchez J. Kisspeptin Stimulates the Pulsatile Secretion of Luteinizing Hormone (LH) during Postpartum Anestrus in Ewes Undergoing Continuous and Restricted Suckling. Animals (Basel) 2021; 11:ani11092656. [PMID: 34573622 PMCID: PMC8469872 DOI: 10.3390/ani11092656] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/07/2021] [Accepted: 08/31/2021] [Indexed: 11/16/2022] Open
Abstract
This study tested whether the intravenous application of kisspeptin can stimulate the pulsatile secretion of LH in suckling ewes during postpartum anestrus. Ten days after lambing, Pelibuey ewes were allocated among two groups: (1) continuous suckling (n = 8), where the lambs remained with their mothers; and (2) restricted suckling (n = 8), where the mothers suckled their lambs twice daily for 30 min. On Day 19 postpartum, the ewes were individually penned with ad libitum access to water and feed and given an indwelling catheter in each jugular vein. On Day 20, 4 mL of blood was sampled every 15 min from 08:00 to 20:00 h to determine LH pulse frequency. At 14:00 h, four ewes in each group received 120 μg of kisspeptin diluted in 3 mL of saline as a continuous infusion for 6 h; the remaining four ewes in each group received only saline. The interaction between kisspeptin and suckling type did not affect LH pulse frequency (p > 0.05). Before kisspeptin administration, pulse frequency was similar in all groups (1.50 ± 0.40 pulses per 6 h; p > 0.05). With the application of kisspeptin, pulse frequency increased to 3.50 ± 0.43 pulses per 6 h (p ≤ 0.014), so the concentration of LH (1.11 ± 0.14 ng mL-1) was greater in kisspeptin-treated ewes than in saline-treated ewes (0.724 ± 0.07 ng mL-1; p ≤ 0.040). The frequency of LH pulses was greater with restricted suckling than with continuous suckling (2.44 ± 0.29 versus 1.69 ± 0.29 pulses per 6 h; p ≤ 0.040). We conclude that intravenous application of kisspeptin increases the pulsatile secretion of LH in suckling ewes and that suckling might reduce kisspeptin neuronal activity, perhaps explaining the suppression of ovulation. Moreover, the effects of kisspeptin and suckling on pulsatile LH secretion appear to be independent, perhaps operating through different neural pathways.
Collapse
Affiliation(s)
- José Manuel Hernández-Hernández
- Programa de Ganadería, Colegio de Postgraduados, Campus Montecillo, km 36.5 Carretera Federal México-Texcoco, Montecillo 56230, Mexico; (J.M.H.-H.); (A.P.-M.)
| | - Graeme B. Martin
- School of Agriculture and Environment, University of Western Australia, Crawley 6009, Australia;
| | | | - Arturo Pro-Martínez
- Programa de Ganadería, Colegio de Postgraduados, Campus Montecillo, km 36.5 Carretera Federal México-Texcoco, Montecillo 56230, Mexico; (J.M.H.-H.); (A.P.-M.)
| | - César Cortez-Romero
- Colegio de Postgraduados, Campus San Luis Potosí, San Luís Potosi 78600, Mexico;
| | - Jaime Gallegos-Sánchez
- Programa de Ganadería, Colegio de Postgraduados, Campus Montecillo, km 36.5 Carretera Federal México-Texcoco, Montecillo 56230, Mexico; (J.M.H.-H.); (A.P.-M.)
- Correspondence: ; Tel.: +52-5959-520-200 (ext. 1723)
| |
Collapse
|
34
|
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: 45] [Impact Index Per Article: 11.3] [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
|
35
|
Xia Q, Chu M, He X, Liu Q, Zhang X, Zhang J, Guo X, Di R. Identification of Photoperiod-Induced LncRNAs and mRNAs in Pituitary Pars Tuberalis of Sheep. Front Vet Sci 2021; 8:644474. [PMID: 34414222 PMCID: PMC8369575 DOI: 10.3389/fvets.2021.644474] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 06/24/2021] [Indexed: 11/13/2022] Open
Abstract
The pituitary pars tuberalis (PT) is the regulating center of seasonal reproduction, which can sense the melatonin signal and eventually cause downstream changes of GnRH secretion through TSHβ. Recently, lncRNAs have been identified in animal reproductive-related tissues, and they play important roles in reproductive regulation. Therefore, in this study, we expect to identify photoperiod-induced lncRNAs and genes in pituitary PT of sheep by comparison of expression profiles between short photoperiod (SP) and long photoperiod (LP). Through RNA-Seq, a total of 55,472 lncRNAs were identified in pituitary PT of Sunite ewes. The number of differentially expressed (DE) genes and lncRNAs between SP and LP increased gradually with the extension of LP (from LP7 to LP42). The notable LP-induced candidate genes included EYA3, TSHB, SIX1, DCT, VMO1, AREG, SUV39H2, and EZH2, and SP-induced genes involved ENSOARG00000012585, CHGA, FOS, SOCS3, and TH. In enriched pathways for DE genes and lncRNA target genes between SP and LP, the reproduction- and circadian-related pathways were highlighted. In addition, the interactome analysis of lncRNAs and their targets implied that MSTRG.209166 and its trans-target TSHB, MSTRG.288068 and its cis-target SIX1, and ENSOARG00000026131 and its cis-target TH might participate in regulation of seasonal reproduction. Together, these results will help to determine important photoperiod-induced lncRNAs and genes and give us some new insights into the epigenetic regulation of seasonal reproduction in sheep.
Collapse
Affiliation(s)
- Qing Xia
- Key Laboratory of Animal Genetics and Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Mingxing Chu
- Key Laboratory of Animal Genetics and Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xiaoyun He
- Key Laboratory of Animal Genetics and Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Qiuyue Liu
- Tianjin Institute of Animal Sciences, Tianjin, China
| | | | - Jinlong Zhang
- Tianjin Institute of Animal Sciences, Tianjin, China
| | - Xiaofei Guo
- Tianjin Institute of Animal Sciences, Tianjin, China
| | - Ran Di
- Key Laboratory of Animal Genetics and Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
36
|
Lindo AN, Thorson JF, Bedenbaugh MN, McCosh RB, Lopez JA, Young SA, Meadows LJ, Bowdridge EC, Fergani C, Freking BA, Lehman MN, Hileman SM, Lents CA. Localization of kisspeptin, NKB, and NK3R in the hypothalamus of gilts treated with the progestin altrenogest. Biol Reprod 2021; 105:1056-1067. [PMID: 34037695 DOI: 10.1093/biolre/ioab103] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 05/17/2021] [Accepted: 05/21/2021] [Indexed: 11/12/2022] Open
Abstract
Mechanisms in the brain controlling secretion of gonadotropin hormones in pigs, particularly luteinizing hormone (LH), are poorly understood. Kisspeptin is a potent LH stimulant that is essential for fertility in many species, including pigs. Neurokinin B (NKB) acting through neurokinin 3 receptor (NK3R) is involved in kisspeptin-stimulated LH release, but organization of NKB and NK3R within the porcine hypothalamus is unknown. Hypothalamic tissue from ovariectomized (OVX) gilts was used to determine the distribution of immunoreactive kisspeptin, NKB, and NK3R cells in the arcuate nucleus (ARC). Almost all kisspeptin neurons coexpressed NKB in the porcine ARC. Immunostaining for NK3R was distributed throughout the preoptic area (POA) and in several hypothalamic areas including the periventricular and retrochiasmatic areas but was not detected within the ARC. There was no colocalization of NK3R with gonadotropin-releasing hormone (GnRH), but NK3R-positive fibers in the POA were in close apposition to GnRH neurons. Treating OVX gilts with the progestin altrenogest decreased LH pulse frequency and reduced mean circulating concentrations of LH compared with OVX control gilts (P < 0.01), but the number of kisspeptin and NKB cells in the ARC did not differ between treatments. The neuroanatomical arrangement of kisspeptin, NKB, and NK3R within the porcine hypothalamus confirm they are positioned to stimulate GnRH and LH secretion in gilts, though differences with other species exist. Altrenogest suppression of LH secretion in the OVX gilt does not appear to involve decreased peptide expression of kisspeptin or NKB.
Collapse
Affiliation(s)
- Ashley N Lindo
- Department of Physiology and Pharmacology and Department of Neuroscience, West Virginia University, Morgantown, WV, USA
| | | | - Michelle N Bedenbaugh
- Department of Physiology and Pharmacology and Department of Neuroscience, West Virginia University, Morgantown, WV, USA
| | - Richard B McCosh
- Department of Physiology and Pharmacology and Department of Neuroscience, West Virginia University, Morgantown, WV, USA
| | - Justin A Lopez
- Department of Physiology and Pharmacology and Department of Neuroscience, West Virginia University, Morgantown, WV, USA
| | - Samantha A Young
- Department of Physiology and Pharmacology and Department of Neuroscience, West Virginia University, Morgantown, WV, USA
| | - Lanny J Meadows
- Department of Physiology and Pharmacology and Department of Neuroscience, West Virginia University, Morgantown, WV, USA
| | - Elizabeth C Bowdridge
- Department of Physiology and Pharmacology and Department of Neuroscience, West Virginia University, Morgantown, WV, USA
| | - Chrysanthi Fergani
- Department of Neurobiology and Anatomical Sciences, The University of Mississippi Medical Center, Jackson, Miss., USA
| | | | - Michael N Lehman
- Department of Biological Sciences and the Brain Health Research Institute, Kent State University, Kent, OH, USA
| | - Stanley M Hileman
- Department of Physiology and Pharmacology and Department of Neuroscience, West Virginia University, Morgantown, WV, USA
| | - Clay A Lents
- USDA, ARS, U.S. Meat Animal Research Center, Clay Center, NE, USA
| |
Collapse
|
37
|
He X, Tao L, Zhong Y, Di R, Xia Q, Wang X, Guo X, Gan S, Zhang X, Zhang J, Liu Q, Chu M. Photoperiod induced the pituitary differential regulation of lncRNAs and mRNAs related to reproduction in sheep. PeerJ 2021; 9:e10953. [PMID: 33976954 PMCID: PMC8067910 DOI: 10.7717/peerj.10953] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 01/27/2021] [Indexed: 12/12/2022] Open
Abstract
The pituitary is a vital endocrine organ that regulates animal seasonal reproduction by controlling the synthesis and secretion of the hormone. The change of photoperiod is the key factor affecting the function of the pituitary in animals, but the mechanism is unclear. Here, we studied the transcriptomic variation in pars distalis (PD) of the pituitary between short photoperiod (SP) and long photoperiod (LP) using RNA sequencing based on the OVX+E2 sheep. 346 differentially expressed (DE) lncRNAs and 186 DE-mRNA were found in the PD. Moreover, function annotation analysis indicated that the reproductive hormones and photoperiod response-related pathways including aldosterone synthesis and secretion, insulin secretion, thyroid hormone synthesis, and circadian entrainment were enriched. The interaction analysis of mRNA-lncRNA suggested that MSTRG.240648, MSTRG.85500, MSTRG.32448, and MSTRG.304959 targeted CREB3L1 and DUSP6, which may be involved in the photoperiodic regulation of the PD. These findings provide resources for further study on the seasonal reproductive in ewes.
Collapse
Affiliation(s)
- Xiaoyun He
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Lin Tao
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yingjie Zhong
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Ran Di
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Qing Xia
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xiangyu Wang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xiaofei Guo
- Tianjin Institute of Animal Sciences, Tianjin, China
| | - Shangquan Gan
- Xinjiang Academy of Agricultural and Reclamation Sciences, Xinjiang, China
| | | | - Jinlong Zhang
- Tianjin Institute of Animal Sciences, Tianjin, China
| | - Qiuyue Liu
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Mingxing Chu
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
38
|
Lakhssassi K, Lahoz B, Sarto P, Iguácel LP, Folch J, Alabart JL, Serrano M, Calvo JH. Genome-Wide Association Study Demonstrates the Role Played by the CD226 Gene in Rasa Aragonesa Sheep Reproductive Seasonality. Animals (Basel) 2021; 11:ani11041171. [PMID: 33921837 PMCID: PMC8074133 DOI: 10.3390/ani11041171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 01/02/2023] Open
Abstract
Simple Summary To elucidate the genetic basis of reproductive seasonality in Rasa Aragonesa sheep breed, we performed a genome-wide association study (GWAS) in order to detect single nucleotide polymorphisms (SNPs) or regions associated with traits related to ovarian function and behavioural signs of estrous. The GWAS included 205 ewes with genotypes for 583882 SNPs. Only one SNP overcame the genome-wide significance level. Nine potential SNPs overcame the chromosome-wise significance level (FDR 10%). Gene annotation demonstrated that CD226molecule (CD226) and neuropeptide Y (NPY) genes that could be involved in reproductive seasonality were close to the significant SNPs. To validate the results, we sequenced the entire coding region of the NPY gene and four exons of the CD226 gene to search for polymorphisms that could be involved in the phenotypes studied. Two synonymous and two nonsynonymous SNPs in the NPY and CD226 genes, respectively, were genotyped in the whole population. We demonstrated that the AA genotype of the SNP rs404360094 located in exon 3 of the CD226 gene was associated with higher and lower total days of anoestrus and oestrous cycling months, respectively. Therefore, this SNP could be utilized as a genetic marker for assisted selection marker to reduce seasonality. Abstract A genome-wide association study (GWAS) was used to identify genomic regions influencing seasonality reproduction traits in Rasa Aragonesa sheep. Three traits associated with either ovarian function based on blood progesterone levels (total days of anoestrus and progesterone cycling months) or behavioral signs of oestrous (oestrous cycling months) were studied. The GWAS included 205 ewes genotyped using the 50k and 680k Illumina Ovine Beadchips. Only one SNP associated with the progesterone cycling months overcame the genome-wide significance level (rs404991855). Nine SNPs exhibited significant associations at the chromosome level, being the SNPs rs404991855 and rs418191944, that are located in the CD226 molecule (CD226) gene, associated with the three traits. This gene is related to reproductive diseases. Two other SNPs were located close to the neuropeptide Y (NPY) gene, which is involved in circadian rhythms. To validate the GWAS, partial characterization of both genes by Sanger sequencing, and genotyping of two synonymous and two nonsynonymous SNPs in the NPY and CD226 genes, respectively, were performed. SNP association analysis showed that only SNP rs404360094 in the exon 3 of the CD226 gene, which produces an amino acid substitution from asparagine (uncharged polar) to aspartic acid (acidic), was associated with the three seasonality traits. Our results suggest that the CD226 gene may be involved in the reproductive seasonality in Rasa Aragonesa.
Collapse
Affiliation(s)
- Kenza Lakhssassi
- Centro de Investigación y Tecnología Agroalimentaria de Aragón, Instituto Agroalimentario de Aragón (IA2) (CITA–Zaragoza University), 50059 Zaragoza, Spain; (K.L.); (B.L.); (P.S.); (L.P.I.); (J.F.); (J.L.A.)
- INRA, Instituts Morocco, 6356 Rabat, Morocco
| | - Belén Lahoz
- Centro de Investigación y Tecnología Agroalimentaria de Aragón, Instituto Agroalimentario de Aragón (IA2) (CITA–Zaragoza University), 50059 Zaragoza, Spain; (K.L.); (B.L.); (P.S.); (L.P.I.); (J.F.); (J.L.A.)
| | - Pilar Sarto
- Centro de Investigación y Tecnología Agroalimentaria de Aragón, Instituto Agroalimentario de Aragón (IA2) (CITA–Zaragoza University), 50059 Zaragoza, Spain; (K.L.); (B.L.); (P.S.); (L.P.I.); (J.F.); (J.L.A.)
| | - Laura Pilar Iguácel
- Centro de Investigación y Tecnología Agroalimentaria de Aragón, Instituto Agroalimentario de Aragón (IA2) (CITA–Zaragoza University), 50059 Zaragoza, Spain; (K.L.); (B.L.); (P.S.); (L.P.I.); (J.F.); (J.L.A.)
| | - José Folch
- Centro de Investigación y Tecnología Agroalimentaria de Aragón, Instituto Agroalimentario de Aragón (IA2) (CITA–Zaragoza University), 50059 Zaragoza, Spain; (K.L.); (B.L.); (P.S.); (L.P.I.); (J.F.); (J.L.A.)
| | - José Luis Alabart
- Centro de Investigación y Tecnología Agroalimentaria de Aragón, Instituto Agroalimentario de Aragón (IA2) (CITA–Zaragoza University), 50059 Zaragoza, Spain; (K.L.); (B.L.); (P.S.); (L.P.I.); (J.F.); (J.L.A.)
| | - Malena Serrano
- Departamento de Mejora Genética Animal INIA, 28040 Madrid, Spain;
| | - Jorge Hugo Calvo
- Centro de Investigación y Tecnología Agroalimentaria de Aragón, Instituto Agroalimentario de Aragón (IA2) (CITA–Zaragoza University), 50059 Zaragoza, Spain; (K.L.); (B.L.); (P.S.); (L.P.I.); (J.F.); (J.L.A.)
- The Aragonese Foundation for Research and Development (ARAID), 50018 Zaragoza, Spain
- Correspondence: ; Tel.: +34976716471
| |
Collapse
|
39
|
Advances in the Regulation of Mammalian Follicle-Stimulating Hormone Secretion. Animals (Basel) 2021; 11:ani11041134. [PMID: 33921032 PMCID: PMC8071398 DOI: 10.3390/ani11041134] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 04/13/2021] [Accepted: 04/13/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary The reproduction of mammals is regulated by the hypothalamic-pituitary-gonadal axis. Follicle stimulating hormone, as one of the gonadotropins secreted by the pituitary gland, plays an immeasurable role. This article mainly reviews the molecular basis and classical signaling pathways that regulate the synthesis and secretion of follicle stimulating hormone, and summarizes its internal molecular mechanism, which provides a certain theoretical basis for the research of mammalian reproduction regulation and the application of follicle stimulating hormone in production practice. Abstract Mammalian reproduction is mainly driven and regulated by the hypothalamic-pituitary-gonadal (HPG) axis. Follicle-stimulating hormone (FSH), which is synthesized and secreted by the anterior pituitary gland, is a key regulator that ultimately affects animal fertility. As a dimeric glycoprotein hormone, the biological specificity of FSH is mainly determined by the β subunit. As research techniques are being continuously innovated, studies are exploring the underlying molecular mechanism regulating the secretion of mammalian FSH. This article will review the current knowledge on the molecular mechanisms and signaling pathways systematically regulating FSH synthesis and will present the latest hypothesis about the nuclear cross-talk among the various endocrine-induced pathways for transcriptional regulation of the FSH β subunit. This article will provide novel ideas and potential targets for the improved use of FSH in livestock breeding and therapeutic development.
Collapse
|
40
|
Liu J, Qu T, Li Z, Yu L, Zhang S, Yuan D, Wu H. Serum kisspeptin levels in polycystic ovary syndrome: A meta-analysis. J Obstet Gynaecol Res 2021; 47:2157-2165. [PMID: 33765692 DOI: 10.1111/jog.14767] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 02/21/2021] [Accepted: 03/10/2021] [Indexed: 11/28/2022]
Abstract
AIM To clarify the association of serum kisspeptin levels in women with polycystic ovary syndrome (PCOS) by meta-analysis. METHODS Two English databases and two Chinese databases were searched for the relationship between kisspeptin and PCOS published from 2009. After the studies screening according to specific principles, we used STATA 12.0 for meta-analysis. Standardized mean difference (SMD) and its 95% confidence intervals (95% CIs) were used as the effect size and STATA 12.0 software was performed by this meta-analysis. RESULTS Nine articles were included in the end, with a total of 1282 participants (699 patients and 583 controls). Heterogeneity between studies was statistically significant. Therefore, the random effects model was used to combine the effects. Meta-analysis showed statistically significant differences in serum kisspeptin levels between the PCOS patients and controls (SMD = 0.57, 95% CI [0.32, 0.82]), which indicated that there is a strong association between serum kisspeptin levels and PCOS. The source of high heterogeneity between the inclusion studies (I2 = 73.2%) might be due to the small sample size. The larger variation of kisspeptin concentration might be caused by different diagnosis criteria of PCOS and short half-time period of kisspeptin combined with nonstandard testing process. CONCLUSION Serum kisspeptin levels in PCOS patients were higher than non-PCOS patients. It is a hint to indicate us that kisspeptin might be an independent biomarker of PCOS patients.
Collapse
Affiliation(s)
- Jing Liu
- Department of Reproductive Medicine, Xi'nan Gynecological Hospital, Chengdu, China
| | - Ting Qu
- Jinxin Research Institute for Reproductive Medicine and Genetics, Chengdu Jinjiang Hospital for Maternal and Child Health Care, Chengdu, China
| | - Zhiyi Li
- Department of Pharmacology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Linlin Yu
- Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Sujuan Zhang
- Jinxin Research Institute for Reproductive Medicine and Genetics, Chengdu Jinjiang Hospital for Maternal and Child Health Care, Chengdu, China
| | - Dongzhi Yuan
- Department of Physiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Haiyan Wu
- Department of Pharmacology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| |
Collapse
|
41
|
Butruille L, Vancamp P, Demeneix BA, Remaud S. Thyroid hormone regulation of adult neural stem cell fate: A comparative analysis between rodents and primates. VITAMINS AND HORMONES 2021; 116:133-192. [PMID: 33752817 DOI: 10.1016/bs.vh.2021.02.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Thyroid hormone (TH) signaling, a highly conserved pathway across vertebrates, is crucial for brain development and function throughout life. In the adult mammalian brain, including that of humans, multipotent neural stem cells (NSCs) proliferate and generate neuronal and glial progenitors. The role of TH has been intensively investigated in the two main neurogenic niches of the adult mouse brain, the subventricular and the subgranular zone. A key finding is that T3, the biologically active form of THs, promotes NSC commitment toward a neuronal fate. In this review, we first discuss the roles of THs in the regulation of adult rodent neurogenesis, as well as how it relates to functional behavior, notably olfaction and cognition. Most research uncovering these roles of TH in adult neurogenesis was conducted in rodents, whose genetic background, brain structure and rate of neurogenesis are considerably different from that of humans. To bridge the phylogenetic gap, we also explore the similarities and divergences of TH-dependent adult neurogenesis in non-human primate models. Lastly, we examine how photoperiodic length changes TH homeostasis, and how that might affect adult neurogenesis in seasonal species to increase fitness. Several aspects by which TH acts on adult NSCs seem to be conserved among mammals, while we only start to uncover the molecular pathways, as well as how other in- and extrinsic factors are intertwined. A multispecies approach delivering more insights in the matter will pave the way for novel NSC-based therapies to combat neurological disorders.
Collapse
Affiliation(s)
- Lucile Butruille
- UMR 7221 Phyma, CNRS/Muséum National d'Histoire Naturelle, Paris, France
| | - Pieter Vancamp
- UMR 7221 Phyma, CNRS/Muséum National d'Histoire Naturelle, Paris, France
| | - Barbara A Demeneix
- UMR 7221 Phyma, CNRS/Muséum National d'Histoire Naturelle, Paris, France
| | - Sylvie Remaud
- UMR 7221 Phyma, CNRS/Muséum National d'Histoire Naturelle, Paris, France.
| |
Collapse
|
42
|
Porter DT, Goodman RL, Hileman SM, Lehman MN. Evidence that synaptic plasticity of glutamatergic inputs onto KNDy neurones during the ovine follicular phase is dependent on increasing levels of oestradiol. J Neuroendocrinol 2021; 33:e12945. [PMID: 33713519 PMCID: PMC7959185 DOI: 10.1111/jne.12945] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 01/12/2021] [Accepted: 01/26/2021] [Indexed: 01/09/2023]
Abstract
Neurones in the arcuate nucleus co-expressing kisspeptin, neurokinin B (NKB) and dynorphin (KNDy) play a critical role in the control of gonadotrophin-releasing hormone (GnRH) and luteinising hormone (LH) secretion. In sheep, KNDy neurones mediate both steroid-negative- and -positive-feedback during pulsatile and preovulatory surge secretions of GnRH/LH, respectively. In addition, KNDy neurones receive glutamatergic inputs expressing vGlut2, a glutamate transporter that serves as a marker for those terminals, from both KNDy neurones and other populations of glutamatergic neurones. Previous work reported higher numbers of vGlut2-positive axonal inputs onto KNDy neurones during the LH surge than in luteal phase ewes. In the present study, we further examined the effects of the ovarian steroids progesterone (P) and oestradiol (E2 ) on glutamatergic inputs to KNDy neurones. Ovariectomised (OVX) ewes received either no further treatment (OVX) or steroid treatments that mimicked the luteal phase (low E2 + P), and early (low E2 ) or late follicular (high E2 ) phases of the oestrous cycle (n = 4 or 5 per group). Brain sections were processed for triple-label immunofluorescent detection of NKB/vGlut2/synaptophysin and analysed using confocal microscopy. We found higher numbers of vGlut2 inputs onto KNDy neurones in high E2 compared to the other three treatment groups. These results suggest that synaptic plasticity of glutamatergic inputs onto KNDy neurones during the ovine follicular phase depend on increasing levels of E2 required for the preovulatory GnRH/surge. These synaptic changes likely contribute to the positive-feedback action of oestrogen on GnRH/LH secretion and thus the generation of the preovulatory surge in the sheep.
Collapse
Affiliation(s)
- Danielle T. Porter
- Neuroscience Graduate Program, Dept. of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center
| | | | | | - Michael N. Lehman
- Brain Health Research Institute and Department of Biological Sciences, Kent State University
- Corresponding author and reprint requests to: Michael N. Lehman, Brain Health Research Institute, Kent State University, 251K Integrated Sciences Building, Kent, Ohio, 44242-0001 USA, Phone: 330-672-2732;
| |
Collapse
|
43
|
Abbara A, Eng PC, Phylactou M, Clarke SA, Richardson R, Sykes CM, Phumsatitpong C, Mills E, Modi M, Izzi-Engbeaya C, Papadopoulou D, Purugganan K, Jayasena CN, Webber L, Salim R, Owen B, Bech P, Comninos AN, McArdle CA, Voliotis M, Tsaneva-Atanasova K, Moenter S, Hanyaloglu A, Dhillo WS. Kisspeptin receptor agonist has therapeutic potential for female reproductive disorders. J Clin Invest 2021; 130:6739-6753. [PMID: 33196464 DOI: 10.1172/jci139681] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 09/03/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUNDKisspeptin is a key regulator of hypothalamic gonadotropin-releasing hormone (GnRH) neurons and is essential for reproductive health. A specific kisspeptin receptor (KISS1R) agonist could significantly expand the potential clinical utility of therapeutics targeting the kisspeptin pathway. Herein, we investigate the effects of a KISS1R agonist, MVT-602, in healthy women and in women with reproductive disorders.METHODSWe conducted in vivo and in vitro studies to characterize the action of MVT-602 in comparison with native kisspeptin-54 (KP54). We determined the pharmacokinetic and pharmacodynamic properties of MVT-602 (doses 0.01 and 0.03 nmol/kg) versus KP54 (9.6 nmol/kg) in the follicular phase of healthy women (n = 9), and in women with polycystic ovary syndrome (PCOS; n = 6) or hypothalamic amenorrhea (HA; n = 6). Further, we investigated their effects on KISS1R-mediated inositol monophosphate (IP1) and Ca2+ signaling in cell lines and on action potential firing of GnRH neurons in brain slices.RESULTSIn healthy women, the amplitude of luteinizing hormone (LH) rise was similar to that after KP54, but peaked later (21.4 vs. 4.7 hours; P = 0.0002), with correspondingly increased AUC of LH exposure (169.0 vs. 38.5 IU∙h/L; P = 0.0058). LH increases following MVT-602 were similar in PCOS and healthy women, but advanced in HA (P = 0.004). In keeping with the clinical data, MVT-602 induced more potent signaling of KISS1R-mediated IP1 accumulation and a longer duration of GnRH neuron firing than KP54 (115 vs. 55 minutes; P = 0.0012).CONCLUSIONTaken together, these clinical and mechanistic data identify MVT-602 as having considerable therapeutic potential for the treatment of female reproductive disorders.TRIAL REGISTRATIONInternational Standard Randomised Controlled Trial Number (ISRCTN) Registry, ISRCTN21681316.FUNDINGNational Institute for Health Research and NIH.
Collapse
Affiliation(s)
- Ali Abbara
- Section of Endocrinology and Investigative Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Pei Chia Eng
- Section of Endocrinology and Investigative Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Maria Phylactou
- Section of Endocrinology and Investigative Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Sophie A Clarke
- Section of Endocrinology and Investigative Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Rachel Richardson
- Institute of Reproductive and Developmental Biology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Charlene M Sykes
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Edouard Mills
- Section of Endocrinology and Investigative Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Manish Modi
- Section of Endocrinology and Investigative Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Chioma Izzi-Engbeaya
- Section of Endocrinology and Investigative Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Debbie Papadopoulou
- Section of Endocrinology and Investigative Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | | | - Channa N Jayasena
- Section of Endocrinology and Investigative Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Lisa Webber
- St Mary's Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom
| | | | - Bryn Owen
- Section of Endocrinology and Investigative Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Paul Bech
- Section of Endocrinology and Investigative Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Alexander N Comninos
- Section of Endocrinology and Investigative Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Craig A McArdle
- Department of Translational Medicine, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | | | - Krasimira Tsaneva-Atanasova
- Department of Mathematics and Living Systems Institute, and.,EPSRC Centre for Predictive Modelling in Healthcare, University of Exeter, Exeter, United Kingdom
| | - Suzanne Moenter
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA.,Department of Internal Medicine, and.,Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA
| | - Aylin Hanyaloglu
- Institute of Reproductive and Developmental Biology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Waljit S Dhillo
- Section of Endocrinology and Investigative Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| |
Collapse
|
44
|
Li Q, Smith JT, Henry B, Rao A, Pereira A, Clarke IJ. Expression of genes for Kisspeptin (KISS1), Neurokinin B (TAC3), Prodynorphin (PDYN), and gonadotropin inhibitory hormone (RFRP) across natural puberty in ewes. Physiol Rep 2021; 8:e14399. [PMID: 32170819 PMCID: PMC7070159 DOI: 10.14814/phy2.14399] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 02/19/2020] [Accepted: 02/21/2020] [Indexed: 01/07/2023] Open
Abstract
Expression of particular genes in hypothami of ewes was measured across the natural pubertal transition by in situ hybridization. The ewes were allocated to three groups (n = 4); prepubertal, postpubertal and postpubertally gonadectomized (GDX). Prepubertal sheep were euthanized at 20 weeks of age and postpubertal animals at 32 weeks. GDX sheep were also euthanized at 32 weeks, 1 week after surgery. Expression of KISS1, TAC3, PDYN in the arcuate nucleus (ARC), RFRP in the dorsomedial hypothalamus and GNRH1 in the preoptic area was quantified on a cellular basis. KISS1R expression by GNRH1 cells was quantified by double-label in situ hybridization. Across puberty, detectable KISS1 cell number increased in the caudal ARC and whilst PDYN cell numbers were low, numbers increased in the rostral ARC. TAC3 expression did not change but RFRP expression/cell was reduced across puberty. There was no change across puberty in the number of GNRH1 cells that expressed the kisspeptin receptor (KISS1R). GDX shortly after puberty did not increase expression of any of the genes of interest. We conclude that KISS1 expression in the ARC increases during puberty in ewes and this may be a causative factor in the pubertal activation of the reproductive axis. A reduction in expression of RFRP may be a factor in the onset of puberty, removing negative tone on GNRH1 cells. The lack of changes in expression of genes following GDX suggest that the effects of gonadal hormones may differ in young and mature animals.
Collapse
Affiliation(s)
- Qun Li
- Department of Physiology, Neuroscience Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Jeremy T Smith
- Department of Physiology, Neuroscience Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Belinda Henry
- Department of Physiology, Neuroscience Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Alexandra Rao
- Department of Physiology, Neuroscience Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Alda Pereira
- Department of Physiology, Neuroscience Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Iain J Clarke
- Department of Physiology, Neuroscience Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
45
|
Uenoyama Y, Nagae M, Tsuchida H, Inoue N, Tsukamura H. Role of KNDy Neurons Expressing Kisspeptin, Neurokinin B, and Dynorphin A as a GnRH Pulse Generator Controlling Mammalian Reproduction. Front Endocrinol (Lausanne) 2021; 12:724632. [PMID: 34566891 PMCID: PMC8458932 DOI: 10.3389/fendo.2021.724632] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 08/23/2021] [Indexed: 01/16/2023] Open
Abstract
Increasing evidence accumulated during the past two decades has demonstrated that the then-novel kisspeptin, which was discovered in 2001, the known neuropeptides neurokinin B and dynorphin A, which were discovered in 1983 and 1979, respectively, and their G-protein-coupled receptors, serve as key molecules that control reproduction in mammals. The present review provides a brief historical background and a summary of our recent understanding of the roles of hypothalamic neurons expressing kisspeptin, neurokinin B, and dynorphin A, referred to as KNDy neurons, in the central mechanism underlying gonadotropin-releasing hormone (GnRH) pulse generation and subsequent tonic gonadotropin release that controls mammalian reproduction.
Collapse
|
46
|
Rumpler É, Skrapits K, Takács S, Göcz B, Trinh SH, Rácz G, Matolcsy A, Kozma Z, Ciofi P, Dhillo WS, Hrabovszky E. Characterization of Kisspeptin Neurons in the Human Rostral Hypothalamus. Neuroendocrinology 2021; 111:249-262. [PMID: 32299085 DOI: 10.1159/000507891] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 04/14/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND Kisspeptin (KP) neurons in the rostral periventricular region of the 3rd ventricle (RP3V) of female rodents mediate positive estrogen feedback to gonadotropin-releasing hormone neurons and, thus, play a fundamental role in the mid-cycle luteinizing hormone (LH) surge. The RP3V is sexually dimorphic, and male rodents with lower KP cell numbers are unable to mount estrogen-induced LH surges. OBJECTIVE To find and characterize the homologous KP neurons in the human brain, we studied formalin-fixed post-mortem hypothalami. METHODS Immunohistochemical techniques were used. RESULTS The distribution of KP neurons in the rostral hypothalamus overlapped with distinct subdivisions of the paraventricular nucleus. The cell numbers decreased after menopause, indicating that estrogens positively regulate KP gene expression in the rostral hypothalamus in humans, similarly to several other species. Young adult women and men had similar cell numbers, as opposed to rodents reported to have more KP neurons in the RP3V of females. Human KP neurons differed from the homologous rodent cells as well, in that they were devoid of enkephalins, galanin and tyrosine hydroxylase. Further, they did not contain known KP neuron markers of the human infundibular nucleus, neurokinin B, substance P and cocaine- and amphetamine-regulated transcript, while they received afferent input from these KP neurons. CONCLUSIONS The identification and positive estrogenic regulation of KP neurons in the human rostral hypothalamus challenge the long-held view that positive estrogen feedback may be restricted to the mediobasal part of the hypothalamus in primates and point to the need of further anatomical, molecular and functional studies of rostral hypothalamic KP neurons.
Collapse
Affiliation(s)
- Éva Rumpler
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Katalin Skrapits
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Szabolcs Takács
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Balázs Göcz
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Sarolta H Trinh
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, 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
| | - Zsolt Kozma
- Department of Forensic Medicine, Faculty of Medicine, University of Pécs, Pécs, Hungary
| | | | - Waljit S Dhillo
- Department of Investigative Medicine, Hammersmith Hospital, Imperial College London, London, United Kingdom
| | - Erik Hrabovszky
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary,
| |
Collapse
|
47
|
Dees WL, Hiney JK, Srivastava VK. IGF-1 Influences Gonadotropin-Releasing Hormone Regulation of Puberty. Neuroendocrinology 2021; 111:1151-1163. [PMID: 33406521 PMCID: PMC8257778 DOI: 10.1159/000514217] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 12/08/2020] [Indexed: 11/19/2022]
Abstract
The pubertal process is initiated as a result of complex neuroendocrine interactions within the preoptic and hypothalamic regions of the brain. These interactions ultimately result in a timely increase in the secretion of gonadotropin-releasing hormone (GnRH). Researchers for years have believed that this increase is due to a diminished inhibitory tone which has applied a prepubertal brake on GnRH secretion, as well as to the gradual development of excitatory inputs driving the increased release of the peptide. Over the years, insulin-like growth factor-1 (IGF-1) has emerged as a prime candidate for playing an important role in the onset of puberty. This review will first present initial research demonstrating that IGF-1 increases in circulation as puberty approaches, is able to induce the release of prepubertal GnRH, and can advance the timing of puberty. More recent findings depict an early action of IGF-1 to activate a pathway that releases the inhibitory brake on prepubertal GnRH secretion provided by dynorphin, as well as demonstrating that IGF-1 can also act later in the process to regulate the synthesis and release of kisspeptin, a potent stimulator of GnRH at puberty.
Collapse
Affiliation(s)
- William L Dees
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, Texas, USA,
| | - Jill K Hiney
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, Texas, USA
| | - Vinod K Srivastava
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
48
|
Hrabovszky E, Takács S, Rumpler É, Skrapits K. The human hypothalamic kisspeptin system: Functional neuroanatomy and clinical perspectives. HANDBOOK OF CLINICAL NEUROLOGY 2021; 180:275-296. [PMID: 34225935 DOI: 10.1016/b978-0-12-820107-7.00017-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In mammals, kisspeptin neurons are the key components of the hypothalamic neuronal networks that regulate the onset of puberty, account for the pulsatile secretion of gonadotropin-releasing hormone (GnRH) and mediate negative and positive estrogen feedback signals to GnRH neurons. Being directly connected anatomically and functionally to the hypophysiotropic GnRH system, the major kisspeptin cell groups of the preoptic area/rostral hypothalamus and the arcuate (or infundibular) nucleus, respectively, are ideally positioned to serve as key nodes which integrate various types of environmental, endocrine, and metabolic signals that can influence fertility. This chapter provides an overview of the current state of knowledge on the anatomy, functions, and plasticity of brain kisspeptin systems based on the wide literature available from different laboratory and domestic species. Then, the species-specific features of human hypothalamic kisspeptin neurons are described, covering their topography, morphology, unique neuropeptide content, plasticity, and connectivity to hypophysiotropic GnRH neurons. Some newly emerging roles of central kisspeptin signaling in behavior and finally, clinical perspectives, are discussed.
Collapse
Affiliation(s)
- Erik Hrabovszky
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary.
| | - Szabolcs Takács
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Éva Rumpler
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Katalin Skrapits
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| |
Collapse
|
49
|
Coen CW, Bennett NC, Holmes MM, Faulkes CG. Neuropeptidergic and Neuroendocrine Systems Underlying Eusociality and the Concomitant Social Regulation of Reproduction in Naked Mole-Rats: A Comparative Approach. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1319:59-103. [PMID: 34424513 DOI: 10.1007/978-3-030-65943-1_3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The African mole-rat family (Bathyergidae) includes the first mammalian species identified as eusocial: naked mole-rats. Comparative studies of eusocial and solitary mole-rat species have identified differences in neuropeptidergic systems that may underlie the phenomenon of eusociality. These differences are found in the oxytocin, vasopressin and corticotrophin-releasing factor (CRF) systems within the nucleus accumbens, amygdala, bed nucleus of the stria terminalis and lateral septal nucleus. As a corollary of their eusociality, most naked mole-rats remain pre-pubertal throughout life because of the presence of the colony's only reproductive female, the queen. To elucidate the neuroendocrine mechanisms that mediate this social regulation of reproduction, research on the hypothalamo-pituitary-gonadal axis in naked mole-rats has identified differences between the many individuals that are reproductively suppressed and the few that are reproductively mature: the queen and her male consorts. These differences involve gonadal steroids, gonadotrophin-releasing hormone-1 (GnRH-1), kisspeptin, gonadotrophin-inhibitory hormone/RFamide-related peptide-3 (GnIH/RFRP-3) and prolactin. The comparative findings in eusocial and solitary mole-rat species are assessed with reference to a broad range of studies on other mammals.
Collapse
Affiliation(s)
- Clive W Coen
- Reproductive Neurobiology, Division of Women's Health, Faculty of Life Sciences & Medicine, King's College London, London, UK.
| | - Nigel C Bennett
- Mammal Research Institute, Department of Zoology and Entomology, University of Pretoria, Pretoria, South Africa
| | - Melissa M Holmes
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON, Canada.,Department of Ecology & Evolutionary Biology, University of Toronto, Toronto, Canada.,Department of Cell & Systems Biology, University of Toronto, Toronto, Canada
| | - Christopher G Faulkes
- School of Biological and Chemical Sciences, Queen Mary University of London, London, UK
| |
Collapse
|
50
|
Beltramo M, Robert V, Decourt C. The kisspeptin system in domestic animals: what we know and what we still need to understand of its role in reproduction. Domest Anim Endocrinol 2020; 73:106466. [PMID: 32247617 DOI: 10.1016/j.domaniend.2020.106466] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 02/10/2020] [Accepted: 02/26/2020] [Indexed: 02/05/2023]
Abstract
The discovery of the kisspeptin (Kp) system stirred a burst of research in the field of reproductive neuroendocrinology. In the last 15 yr, the organization and activity of the system, including its neuroanatomical structure, its major physiological functions, and its main pharmacological properties, were outlined. To this endeavor, the use of genetic tools to delete and to restore Kp system functionality in a specific tissue was essential. At present, there is no question as to the key role of the Kp system in mammalian reproduction. However, easily applicable genetic manipulations are unavailable for domestic animals. Hence, many essential details on the physiological mechanisms underlying its action on domestic animals require further investigation. The potentially different effects of the various Kp isoforms, the precise anatomical localization of the Kp receptor, and the respective role played by the 2 main populations of Kp cells in different species are only few of the questions that remain unanswered and that will be illustrated in this review. Furthermore, the application of synthetic pharmacologic tools to manipulate the Kp system is still in its infancy but has produced some interesting results, suggesting the possibility of developing new methods to manage reproduction in domestic animals. In spite of a decade and a half of intense research effort, much work is still required to achieve a comprehensive understanding of the influence of the Kp system on reproduction. Furthermore, Kp system ramifications in other physiological functions are emerging and open new research perspectives.
Collapse
Affiliation(s)
- M Beltramo
- INRAE (CNRS, UMR7247, Université de Tours, IFCE), UMR85 Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, France.
| | - V Robert
- INRAE (CNRS, UMR7247, Université de Tours, IFCE), UMR85 Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, France
| | - C Decourt
- INRAE (CNRS, UMR7247, Université de Tours, IFCE), UMR85 Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, France
| |
Collapse
|