1
|
Rousselle D, Silveyra P. Acute Exposure to Ozone Affects Circulating Estradiol Levels and Gonadotropin Gene Expression in Female Mice. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2025; 22:222. [PMID: 40003448 PMCID: PMC11855596 DOI: 10.3390/ijerph22020222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 01/29/2025] [Accepted: 02/03/2025] [Indexed: 02/27/2025]
Abstract
Ozone, a critical air pollutant, has been shown to lead to systemic inflammation that can alter bodily functions, including hormone secretion, fertility, and the hypothalamic-pituitary-gonadal (HPG) axis. This study aimed to quantify changes in hormone production and follicle development after acute exposure to ozone using an animal model to identify the potential mechanisms underlying the observed effects of air pollution exposures on fertility and hormone secretion. To accomplish this, regularly cycling 8-week-old female C57BL/6J mice were exposed to 2 ppm of ozone or filtered air (control) for 3 h on the day of proestrus. Blood, ovaries, brain tissues, and pituitary glands were collected at 4 h after exposure to evaluate hormone levels, ovarian follicle distribution, and gene expression. Ovaries were also harvested at 24 h post-exposure. We found that at 4 h after ozone exposure, mice had significantly higher (30%) circulating estradiol levels than mice exposed to filtered air. This effect was accompanied by a decrease in mRNA expression of gonadotropin genes (LH, FSH) and gonadotropin-releasing hormone in the pituitary gland. Analysis of ovarian tissue at 4 h and 24 h after exposure showed no significant changes in follicle composition or the expression of steroidogenesis genes. We conclude that acute ozone exposure affects sex hormone levels and disrupts the HPG axis. Future studies addressing chronic or long-term effects of air pollution exposure are needed to elucidate the mechanisms by which ambient ozone affects endocrine function.
Collapse
Affiliation(s)
- Dustin Rousselle
- Department of Environmental and Occupational Health, School of Public Health, Indiana University, Bloomington, IN 47405, USA;
| | - Patricia Silveyra
- Department of Environmental and Occupational Health, School of Public Health, Indiana University, Bloomington, IN 47405, USA;
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
2
|
Göcz B, Rumpler É, Szentkirályi-Tóth S, Skrapits K, Takács S, Sárvári M, Farkas I, Póliska S, Hrabovszky E. Laser-capture microdissection for spatial transcriptomics of immunohistochemically detected neurons. J Biol Chem 2025; 301:108150. [PMID: 39736395 PMCID: PMC11910328 DOI: 10.1016/j.jbc.2024.108150] [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: 09/22/2024] [Revised: 11/29/2024] [Accepted: 12/25/2024] [Indexed: 01/01/2025] Open
Abstract
We developed a versatile 'IHC/LCM-Seq' method for spatial transcriptomics of immunohistochemically detected neurons collected with laser-capture microdissection (LCM). IHC/LCM-Seq uses aluminon and polyvinyl sulfonic acid for inventive RNA-preserving strategies to maintain RNA integrity in free-floating sections of 4% formaldehyde-fixed brains. To validate IHC/LCM-Seq, we first immunostained and harvested striatal cholinergic interneurons with LCM. RNA preparations were subjected to random primer-based cDNA library preparation and bulk sequencing on the NextSeq Illumina platform. IHC/LCM-Seq detected ∼16,000 transcripts, reaching the sensitivity of a reference 'LCM-Seq method' developed for fluorescently tagged neurons microdissected from lightly formaldehyde-fixed and slide-mounted brain sections of transgenic mice. We successfully used the new IHC/LCM-Seq approach to provide unprecedented insight into the transcriptome of immunohistochemically detected gonadotropin-releasing hormone (GnRH) neurons regulating reproduction. The ∼13,000 to 14,000 transcripts identified in GnRH neurons of adult male rats and mice encoded 28 proteins implicated previously in human infertility, 35 neuropeptides, 34 nuclear receptors, and 164 G protein-coupled receptors. Functional experiments using slice electrophysiology established that the heavy Ntsr2 expression conveys a strong excitatory action of neurotensin on GnRH neurons. As an unexpected species difference, we found that GnRH neurons exclusively expressed estrogen receptor-β in rats and against the current consensus, the alpha estrogen receptor isoform in mice. The IHC/LCM-Seq technique we are reporting is a highly sensitive and accurate bulk sequencing approach to characterize the transcriptome landscape of immunohistochemically labeled neurons, including neuroendocrine GnRH cells. This method is readily applicable to any species, opening new perspectives also for future studies of the post mortem human brain.
Collapse
Affiliation(s)
- Balázs Göcz
- Laboratory of Reproductive Neurobiology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary.
| | - Éva Rumpler
- Laboratory of Reproductive Neurobiology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary.
| | - Soma Szentkirályi-Tóth
- Laboratory of Reproductive Neurobiology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Katalin Skrapits
- Laboratory of Reproductive Neurobiology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Szabolcs Takács
- Laboratory of Reproductive Neurobiology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Miklós Sárvári
- Laboratory of Reproductive Neurobiology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Imre Farkas
- Laboratory of Reproductive Neurobiology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Szilárd Póliska
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Erik Hrabovszky
- Laboratory of Reproductive Neurobiology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary.
| |
Collapse
|
3
|
Leng D, Zeng B, Wang T, Chen BL, Li DY, Li ZJ. Single nucleus/cell RNA-seq of the chicken hypothalamic-pituitary-ovarian axis offers new insights into the molecular regulatory mechanisms of ovarian development. Zool Res 2024; 45:1088-1107. [PMID: 39245652 PMCID: PMC11491784 DOI: 10.24272/j.issn.2095-8137.2024.037] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 06/17/2024] [Indexed: 09/10/2024] Open
Abstract
The hypothalamic-pituitary-ovarian (HPO) axis represents a central neuroendocrine network essential for reproductive function. Despite its critical role, the intrinsic heterogeneity within the HPO axis across vertebrates and the complex intercellular interactions remain poorly defined. This study provides the first comprehensive, unbiased, cell type-specific molecular profiling of all three components of the HPO axis in adult Lohmann layers and Liangshan Yanying chickens. Within the hypothalamus, pituitary, and ovary, seven, 12, and 13 distinct cell types were identified, respectively. Results indicated that the pituitary adenylate cyclase activating polypeptide (PACAP), follicle-stimulating hormone (FSH), and prolactin (PRL) signaling pathways may modulate the synthesis and secretion of gonadotropin-releasing hormone (GnRH), FSH, and luteinizing hormone (LH) within the hypothalamus and pituitary. In the ovary, interactions between granulosa cells and oocytes involved the KIT, CD99, LIFR, FN1, and ANGPTL signaling pathways, which collectively regulate follicular maturation. The SEMA4 signaling pathway emerged as a critical mediator across all three tissues of the HPO axis. Additionally, gene expression analysis revealed that relaxin 3 (RLN3), gastrin-releasing peptide (GRP), and cocaine- and amphetamine regulated transcripts (CART, also known as CARTPT) may function as novel endocrine hormones, influencing the HPO axis through autocrine, paracrine, and endocrine pathways. Comparative analyses between Lohmann layers and Liangshan Yanying chickens demonstrated higher expression levels of GRP, RLN3, CARTPT, LHCGR, FSHR, and GRPR in the ovaries of Lohmann layers, potentially contributing to their superior reproductive performance. In conclusion, this study provides a detailed molecular characterization of the HPO axis, offering novel insights into the regulatory mechanisms underlying reproductive biology.
Collapse
Affiliation(s)
- Dong Leng
- School of Pharmacy, Chengdu University, Chengdu, Sichuan 610106, China
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Bo Zeng
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Tao Wang
- School of Pharmacy, Chengdu University, Chengdu, Sichuan 610106, China
| | - Bin-Long Chen
- College of Animal Science, Xichang University, Xichang, Sichuan 615000, China. E-mail:
| | - Di-Yan Li
- School of Pharmacy, Chengdu University, Chengdu, Sichuan 610106, China. E-mail:
| | - Zhuan-Jian Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan 450046, China. E-mail:
| |
Collapse
|
4
|
Bhattacharya K, Dey R, Sen D, Paul N, Basak AK, Purkait MP, Shukla N, Chaudhuri GR, Bhattacharya A, Maiti R, Adhikary K, Chatterjee P, Karak P, Syamal AK. Polycystic ovary syndrome and its management: In view of oxidative stress. Biomol Concepts 2024; 15:bmc-2022-0038. [PMID: 38242137 DOI: 10.1515/bmc-2022-0038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/11/2023] [Indexed: 01/21/2024] Open
Abstract
In the past two decades, oxidative stress (OS) has drawn a lot of interest due to the revelation that individuals with many persistent disorders including diabetes, polycystic ovarian syndrome (PCOS), cardiovascular, and other disorders often have aberrant oxidation statuses. OS has a close interplay with PCOS features such as insulin resistance, hyperandrogenism, and chronic inflammation; there is a belief that OS might contribute to the development of PCOS. PCOS is currently recognized as not only one of the most prevalent endocrine disorders but also a significant contributor to female infertility, affecting a considerable proportion of women globally. Therefore, the understanding of the relationship between OS and PCOS is crucial to the development of therapeutic and preventive strategies for PCOS. Moreover, the mechanistic study of intracellular reactive oxygen species/ reactive nitrogen species formation and its possible interaction with women's reproductive health is required, which includes complex enzymatic and non-enzymatic antioxidant systems. Apart from that, our current review includes possible regulation of the pathogenesis of OS. A change in lifestyle, including physical activity, various supplements that boost antioxidant levels, particularly vitamins, and the usage of medicinal herbs, is thought to be the best way to combat this occurrence of OS and improve the pathophysiologic conditions associated with PCOS.
Collapse
Affiliation(s)
- Koushik Bhattacharya
- School of Paramedics and Allied Health Sciences, Centurion University of Technology and Management, Khurda Road, Bhubaneswar, Odisha, India
| | - Rajen Dey
- Department of Medical Laboratory Technology, Swami Vivekananda University, Barrackpore, West Bengal, India
| | - Debanjana Sen
- Post-Graduate Department of Physiology, Hooghly Mohsin College, Chinsurah, West-Bengal, India
| | - Nimisha Paul
- Department of General Human Physiology and Biochemistry, Hitkarini Dental College and Hospital, Jabalpur, Madhya Pradesh, India
| | - Asim Kumar Basak
- School of Allied Health Sciences, Brainware University, Barasat, West-Bengal, India
| | | | - Nandini Shukla
- Department of Anatomy, Pt. J.N.M. Medical College, Raipur, Chhattisgarh, India
| | - Gargi Ray Chaudhuri
- Department of Physiotherapy, Nopany Institute of Health Care Studies, Kolkata, West-Bengal, India
| | - Aniruddha Bhattacharya
- Department of Physiology, International Medical School, Management and Science University, Selangor, Malaysia
| | - Rajkumar Maiti
- Department of Physiology, Bankura Christian College, Bankura, West Bengal, India
| | - Krishnendu Adhikary
- Department of Interdisciplinary Science, Centurion University of Technology and Management, Khurda Road, Bhubaneswar, Odisha, India
| | - Prity Chatterjee
- Department of Biotechnology, Paramedical College, Durgapur, West Bengal, India
| | - Prithviraj Karak
- Department of Physiology, Bankura Christian College, Bankura, West Bengal, India
| | - Alak Kumar Syamal
- Post-Graduate Department of Physiology, Hooghly Mohsin College, Chinsurah, West-Bengal, India
| |
Collapse
|
5
|
Zimerman J, Niño OMS, da Costa CS, Zanol JF, Comério M, da Gama de Souza LN, Miranda-Alves L, Miranda RA, Lisboa PC, Camilo TA, Rorato R, Alves GA, Frazão R, Zomer HD, Freitas-Lima LC, Graceli JB. Subacute high-refined carbohydrate diet leads to abnormal reproductive control of the hypothalamic-pituitary axis in female rats. Reprod Toxicol 2023; 119:108410. [PMID: 37211340 DOI: 10.1016/j.reprotox.2023.108410] [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: 03/12/2023] [Revised: 05/05/2023] [Accepted: 05/18/2023] [Indexed: 05/23/2023]
Abstract
We previously reported that female rats placed on a diet containing refined carbohydrates (HCD) resulted in obesity and reproductive abnormalities, such as high serum LH concentration and abnormal ovarian function. However, the impacts at the hypothalamic-pituitary (HP) function, specifically regarding pathways linked to reproductive axis modulation are unknown. In this study, we assessed whether subacute feeding with HCD results in abnormal reproductive control in the HP axis. Female rats were fed with HCD for 15 days and reproductive HP axis morphophysiology was assessed. HCD reduced hypothalamic mRNA expression (Kiss1, Lepr, and Amhr2) and increased pituitary LHβ+ cells. These changes likely contribute to the increase in serum LH concentration observed in HCD. Blunted estrogen negative feedback was observed in HCD, with increased kisspeptin protein expression in the arcuate nucleus of the hypothalamus (ARH), lower LHβ+ cells and LH concentration in ovariectomized (OVX)+HCD rats. Thus, these data suggest that HCD feeding led to female abnormal reproductive control of HP axis.
Collapse
Affiliation(s)
- Jeanini Zimerman
- Department of Morphology, Federal University of Espírito Santo, Vitória, Brazil
| | - Oscar M S Niño
- Department of Morphology, Federal University of Espírito Santo, Vitória, Brazil; Faculty of Human Sciences and Education, Universidad de los Llanos, Villavicencio, Meta, Colombia
| | - Charles S da Costa
- Department of Morphology, Federal University of Espírito Santo, Vitória, Brazil
| | - Jordana F Zanol
- Department of Morphology, Federal University of Espírito Santo, Vitória, Brazil
| | - Milena Comério
- Department of Morphology, Federal University of Espírito Santo, Vitória, Brazil
| | | | - Leandro Miranda-Alves
- Experimental Endocrinology Research, Development and Innovation Group, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Brazil; Postgraduate Program in Endocrinology, School of Medicine, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho, Ilha do Governador, Cidade Universitária, UFRJ, RJ, Brazil
| | - Rosiane A Miranda
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, RJ, Brazil
| | - Patrícia C Lisboa
- Laboratory of Endocrine Physiology, Biology Institute, State University of Rio de Janeiro, RJ, Brazil
| | - Tays A Camilo
- Department of Biophysics, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Rodrigo Rorato
- Department of Biophysics, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Guilherme Andrade Alves
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Renata Frazão
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Helena D Zomer
- Department of Physiological Sciences, University of Florida, Gainesville, FL, USA
| | | | - Jones B Graceli
- Department of Morphology, Federal University of Espírito Santo, Vitória, Brazil.
| |
Collapse
|
6
|
Functional Characterization of Three GnRH Isoforms in Small Yellow Croaker Larimichthys polyactis Maintained in Captivity: Special Emphasis on Reproductive Dysfunction. BIOLOGY 2022; 11:biology11081200. [PMID: 36009826 PMCID: PMC9404844 DOI: 10.3390/biology11081200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/07/2022] [Accepted: 08/09/2022] [Indexed: 11/16/2022]
Abstract
Fish reproduction is regulated by the brain–pituitary–gonad (BPG) axis where the gonadotropin-releasing hormone (GnRH) plays a central role. Seed production of small yellow croaker (Larimichthys polyactis) is performed using captive-reared broodstock known to undergo reproductive dysfunction, which is connected to endocrinological dysfunction. To determine the endocrinological mechanism of GnRHs in the BPG axis of small yellow croaker, full-length sequences of three GnRH isoforms encoding sbGnRH (GnRH1), cGnRH-II (GnRH2), and sGnRH (GnRH3) were cloned and characterized from brain tissue. qRT-PCR, in vivo, and in vitro experiments were performed for functional characterization. The mRNA expression of GnRH1 in the brain and gonadotropin subunits (GPα, FSHβ, and LHβ) in the pituitary were significantly higher at the ripen stage during gonadal development and GnRH1 at spawning stage during spawning events. Expression of both GnRH1 and GtH subunits was significantly lower in females than males. GtH subunits were induced at higher concentrations of GnRH1 in vivo and in vitro. Sex-steroids significantly inhibited the GnRH1 expression in vitro in a dose-dependent manner. Taken together, results indicated that GnRH1 plays a key role in gonadal maturation and sex-steroids induced negative feedback in the regulation of GnRH. A lower level of GnRH1 and GtHs might be responsible for reproductive dysfunction in a female small yellow croaker.
Collapse
|
7
|
GABAergic and Glutamatergic Phenotypes of Neurons Expressing Calcium-Binding Proteins in the Preoptic Area of the Guinea Pig. Int J Mol Sci 2022; 23:ijms23147963. [PMID: 35887305 PMCID: PMC9320123 DOI: 10.3390/ijms23147963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/18/2022] [Accepted: 07/18/2022] [Indexed: 11/17/2022] Open
Abstract
The mammalian preoptic area (POA) has large populations of calbindin (CB), calretinin (CR) and parvalbumin (PV) neurons, but phenotypes of these cells are unknown. Therefore, the question is whether neurons expressing CB, CR, and/or PV are GABAergic or glutamatergic. Double-immunofluorescence staining followed by epifluorescence and confocal microscopy was used to determine the coexpression patterns of CB, CR and PV expressing neurons with vesicular GABA transporters (VGAT) as specific markers of GABAergic neurons and vesicular glutamate transporters (VGLUT 2) as specific markers of glutamatergic neurons. The guinea pig was adopted as, like humans, it has a reproductive cycle with a true luteal phase and a long gestation period. The results demonstrated that in the guinea pig POA of both sexes, ~80% of CB+ and ~90% of CR+ neurons coexpress VGAT; however, one-fifth of CB+ neurons and one-third of CR+ cells coexpress VGLUT. About two-thirds of PV+ neurons express VGAT, and similar proportion of them coexpress VGLUT. Thus, many CB+, CR+ and PV+ neurons may be exclusively GABAergic (VGAT-expressing cells) or glutamatergic (VGLUT-expressing cells); however, at least a small fraction of CR+ cells and at least one-third of PV+ cells are likely neurons with a dual GABA/glutamate phenotype that may coexpress both transporters.
Collapse
|
8
|
Grant CV, Russart KLG, Pyter LM. A novel targeted approach to delineate a role for estrogen receptor-β in ameliorating murine mammary tumor-associated neuroinflammation. Endocrine 2022; 75:949-958. [PMID: 34797509 DOI: 10.1007/s12020-021-02931-7] [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: 08/17/2021] [Accepted: 10/27/2021] [Indexed: 11/28/2022]
Abstract
PURPOSE Circulating estrogens in breast cancer patients and survivors are often extremely low due to menopause and estrogen-reducing cancer treatments. Simultaneously, circulating inflammatory markers, and inflammatory proteins in brains of rodent tumor models, can be elevated and correlate with debilitating neurological and psychological comorbidities. Because estrogen has anti-inflammatory properties in the brain, we hypothesized that mammary tumor-induced neuroinflammation is driven, in part, by reduced brain estrogen signaling. METHODS An ovariectomized mouse model of postmenopausal breast cancer utilizing the ERα-positive 67NR mammary tumor cell line was used for these experiments. A novel, orally bioavailable, and brain penetrant ERβ agonist was administered daily via oral gavage. Following treatment, estrogen-responsive genes were measured in brain regions. Central and circulating inflammatory markers were measured via RT-qPCR and a multiplex cytokine array, respectively. RESULTS We present novel findings that peripheral mammary tumors alter estrogen signaling genes including receptors and aromatase in the hypothalamus, hippocampus, and frontal cortex. Mammary tumors induced peripheral and central inflammation, however, pharmacological ERβ activation was not sufficient to reduce this inflammation. CONCLUSIONS Data presented here suggest that compensating for low circulating estrogen with ERβ brain activation is not sufficient to attenuate mammary tumor-induced neuroinflammation, and is therefore not a likely candidate for the treatment of behavioral symptoms in patients. The novel finding that mammary tumors alter estrogen signaling-related genes is a clinically relevant advancement to the understanding of how peripheral tumor biology modulates neurobiology. This is necessary to predict and prevent behavioral comorbidities (e.g., cognitive impairment) prevalent in cancer patients and survivors.
Collapse
Affiliation(s)
- Corena V Grant
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
- Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, OH, USA
| | - Kathryn L G Russart
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Minnesota State University Moorhead, Moorhead, MN, USA
| | - Leah M Pyter
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA.
- Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, OH, USA.
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
9
|
Faykoo-Martinez M, Kalinowski LM, Holmes MM. Neuroendocrine regulation of pubertal suppression in the naked mole-rat: What we know and what comes next. Mol Cell Endocrinol 2021; 534:111360. [PMID: 34116130 DOI: 10.1016/j.mce.2021.111360] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/20/2021] [Accepted: 06/03/2021] [Indexed: 01/13/2023]
Abstract
Puberty is a key developmental milestone that marks an individual's maturation in several ways including, but not limited to, reproductive maturation, changes in behaviors and neural organization. The timing at which puberty occurs is variable both within individuals of the same species and between species. These variations can be aligned with ecological cues that delay or suppress puberty. Naked mole-rats are colony-living rodents where reproduction is restricted to a few animals; all other animals are pubertally-suppressed. Animals removed from suppressive colony cues can reproductively mature, presenting the unique opportunity to study adult-onset puberty. Recently, we found that RFRP-3 administration sustains pubertal delay in naked mole-rats removed from colony. In this review, we explore what is known about regulators that control puberty onset, the role of stress/social status in pubertal timing, the status of knowledge of pubertal suppression in naked mole-rats and what comes next.
Collapse
Affiliation(s)
| | | | - Melissa M Holmes
- Department of Cell and Systems Biology, University of Toronto, Canada; Department of Psychology, University of Toronto Mississauga, Canada; Department of Ecology and Evolutionary Biology, University of Toronto, Canada
| |
Collapse
|
10
|
Ma Y, Shi Y, Wu Q, Ma W. Epigallocatechin-3-gallate Alleviates Vanadium-Induced Reduction of Antioxidant Capacity via Keap1-Nrf2-sMaf Pathway in the Liver, Kidney, and Ovary of Laying Hens. Biol Trace Elem Res 2021; 199:2707-2716. [PMID: 33405082 DOI: 10.1007/s12011-020-02398-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/14/2020] [Indexed: 11/29/2022]
Abstract
This study evaluated the effect of epigallocatechin-3-gallate (EGCG) alleviating the reduction of antioxidant capacity induced by dietary vanadium (V) in the liver, kidney, and ovary of laying hens. Furthermore, Kelch-like ECH-associated protein 1(Keap1)-nuclear factor erythroid 2-related factor 2 (Nrf2)-small Maf proteins (sMaf) pathway was explored to reveal the molecular mechanism. A total of 768 40-week-old Hyline-Brown laying hens were randomly allocated to 4 groups with 8 pens per group and 24 hens per pen. The experimental groups were as follows: control (basal diet); V15, control + 15 mg/kg V; EGCG150, control + 150 mg/kg EGCG; V15 + EGCG150, V15 + 150 mg/kg EGCG. Our results revealed that dietary EGCG supplementation completely alleviated the V-induced reductions of hen-day egg production, average egg weight, Haugh unit, albumen height, eggshell strength, and eggshell thickness. Dietary EGCG supplementation completely prevented the V-induced reductions of serum follicle-stimulating hormone and luteinizing hormone levels. Besides, dietary EGCG supplementation reversed the V-induced increments of alanine aminotransferase (ALT), aspartate aminotransferase (AST), blood urea nitrogen (BUN), creatinine (Cr), and uric acid (UA). In addition, dietary EGCG supplementation partially alleviated the V-induced reductions of the enzyme activities and gene expressions of superoxidative dismutase (SOD), catalase (CAT), glutathione reductase (GR), and glutathione peroxidase (GSH-Px). Furthermore, dietary EGCG supplementation partially alleviated the V-induced reductions of Nrf2 and sMaf gene expressions, and the increments of Keap1 gene expression. In summary, EGCG partially alleviated V-induced reduction of antioxidant capacity through Keap1-Nrf2-sMaf pathway in the liver, kidney, and ovary of laying hens.
Collapse
Affiliation(s)
- Yan Ma
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471003, China.
| | - Yizhen Shi
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471003, China
| | - Qiujue Wu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471003, China
| | - Wenfeng Ma
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471003, China
| |
Collapse
|
11
|
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
|
12
|
Naulé L, Maione L, Kaiser UB. Puberty, A Sensitive Window of Hypothalamic Development and Plasticity. Endocrinology 2021; 162:bqaa209. [PMID: 33175140 PMCID: PMC7733306 DOI: 10.1210/endocr/bqaa209] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Indexed: 12/12/2022]
Abstract
Puberty is a developmental period characterized by a broad range of physiologic changes necessary for the acquisition of adult sexual and reproductive maturity. These changes mirror complex modifications within the central nervous system, including within the hypothalamus. These modifications result in the maturation of a fully active hypothalamic-pituitary-gonadal (HPG) axis, the neuroendocrine cascade ensuring gonadal activation, sex steroid secretion, and gametogenesis. A complex and finely regulated neural network overseeing the HPG axis, particularly the pubertal reactivation of gonadotropin-releasing hormone (GnRH) secretion, has been progressively unveiled in the last 3 decades. This network includes kisspeptin, neurokinin B, GABAergic, and glutamatergic neurons as well as glial cells. In addition to substantial modifications in the expression of key targets, several changes in neuronal morphology, neural connections, and synapse organization occur to establish mature and coordinated neurohormonal secretion, leading to puberty initiation. The aim of this review is to outline the current knowledge of the major changes that neurons secreting GnRH and their neuronal and glial partners undergo before and after puberty. Emerging mediators upstream of GnRH, uncovered in recent years, are also addressed herein. In addition, the effects of sex steroids, particularly estradiol, on changes in hypothalamic neurodevelopment and plasticity are discussed.
Collapse
Affiliation(s)
- Lydie Naulé
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Luigi Maione
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Paris Saclay University, Assistance Publique-Hôpitaux de Paris, Department Endocrinology and Reproductive Diseases, Bicêtre Hospital, Paris, France
| | - Ursula B Kaiser
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
13
|
Teo CH, Phon B, Parhar I. The Role of GnIH in Biological Rhythms and Social Behaviors. Front Endocrinol (Lausanne) 2021; 12:728862. [PMID: 34566893 PMCID: PMC8461181 DOI: 10.3389/fendo.2021.728862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/23/2021] [Indexed: 12/30/2022] Open
Abstract
Gonadotropin-inhibitory hormone (GnIH) was first discovered in the Japanese quail, and peptides with a C-terminal LPXRFamide sequence, the signature protein structure defining GnIH orthologs, are well conserved across vertebrate species, including fish, reptiles, amphibians, avians, and mammals. In the mammalian brain, three RFamide-related proteins (RFRP-1, RFRP-2, RFRP-3 = GnIH) have been identified as orthologs to the avian GnIH. GnIH is found primarily in the hypothalamus of all vertebrate species, while its receptors are distributed throughout the brain including the hypothalamus and the pituitary. The primary role of GnIH as an inhibitor of gonadotropin-releasing hormone (GnRH) and pituitary gonadotropin release is well conserved in mammalian and non-mammalian species. Circadian rhythmicity of GnIH, regulated by light and seasons, can influence reproductive activity, mating behavior, aggressive behavior, and feeding behavior. There is a potential link between circadian rhythms of GnIH, anxiety-like behavior, sleep, stress, and infertility. Therefore, in this review, we highlight the functions of GnIH in biological rhythms, social behaviors, and reproductive and non-reproductive activities across a variety of mammalian and non-mammalian vertebrate species.
Collapse
|
14
|
Khodamoradi K, Khosravizadeh Z, Amini-Khoei H, Hosseini SR, Dehpour AR, Hassanzadeh G. The effects of maternal separation stress experienced by parents on male reproductive potential in the next generation. Heliyon 2020; 6:e04807. [PMID: 33024852 PMCID: PMC7527646 DOI: 10.1016/j.heliyon.2020.e04807] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 05/31/2020] [Accepted: 08/25/2020] [Indexed: 11/17/2022] Open
Abstract
There is little information available about the effects of early-life parental stress on the reproductive potential of the next generation. The aim of this study is to examine the reproductive potential of male mice whose parents experienced maternal separation stress. In the present study, male first-generation offspring from parents were undergone of maternal separation (MS) were examined. Sperm characteristics, histological changes in testis, reactive oxygen species (ROS) production, expression of apoptotic and inflammatory genes and proteins were assessed. Findings showed that MS experienced by parents significantly decreased the morphology and viability of spermatozoa. Furthermore, significant changes in testicular tissue histology were observed. Increased production of ROS, decreased glutathione peroxidase (GPX) and adenosine triphosphate (ATP) concentrations, and affected the expression of genes and cytokines involved in inflammation. Finally, the mean percentage of caspase-1 and NLRP3 (NOD-, LRR- and pyrin domain-containing protein 3) positive cells was significantly higher in first-generation group. MS experienced by parents may negatively affect the reproduction of first generation offspring.
Collapse
Affiliation(s)
- Kajal Khodamoradi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Zahra Khosravizadeh
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Amini-Khoei
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Seyed Reza Hosseini
- Departent of Urology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Gholamreza Hassanzadeh
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Li S, Zhai J, Xu B, Liu J, Chu W, Wang D, Geng X, Chen ZJ, Du Y. Erythropoietin-producing hepatocellular receptor A7 restrains estrogen negative feedback of luteinizing hormone via ephrin A5 in the hypothalamus of female rats. Am J Physiol Endocrinol Metab 2020; 319:E81-E90. [PMID: 32396496 DOI: 10.1152/ajpendo.00046.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
We have previously shown that systemic injection of erythropoietin-producing hepatocellular receptor A7 (EPHA7)-Fc raises serum luteinizing hormone (LH) levels before ovulation in female rats, indicating the induction of EPHA7 in ovulation. In this study, we aimed to identify the mechanism and hypothalamus-pituitary-ovary (HPO) axis level underlying the promotion of LH secretion by EPHA7. Using an ovariectomized (OVX) rat model, in conjunction with low-dose 17β-estradiol (E2) treatment, we investigated the association between EPHA7-ephrin (EFN)A5 signaling and E2 negative feedback. Various rat models (OVX, E2-treated OVX, and abarelix treated) were injected with the recombinant EPHA7-Fc protein through the caudal vein to investigate the molecular mechanism underlying the promotion of LH secretion by EPHA7. Efna5 was observed strongly expressed in the arcuate nucleus of the female rat by using RNAscope in situ hybridization. Our results indicated that E2, combined with estrogen receptor (ER)α, but not ERβ, inhibited Efna5 and gonadotropin-releasing hormone 1 (Gnrh1) expressions in the hypothalamus. In addition, the systemic administration of EPHA7-Fc restrained the inhibition of Efna5 and Gnrh1 by E2, resulting in increased Efna5 and Gnrh1 expressions in the hypothalamus as well as increased serum LH levels. Collectively, our findings demonstrated the involvement of EPHA7-EFNA5 signaling in the regulation of LH and the E2 negative feedback pathway in the hypothalamus, highlighting the functional role of EPHA7 in female reproduction.
Collapse
Affiliation(s)
- Shang Li
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai, Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Junyu Zhai
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai, Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Bing Xu
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai, Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Jiansheng Liu
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai, Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Weiwei Chu
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai, Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Dongshuang Wang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai, Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Xueying Geng
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai, Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai, Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
- Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, China
| | - Yanzhi Du
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai, Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| |
Collapse
|
16
|
Single-Cell Gene Profiling Reveals Social Status-Dependent Modulation of Nuclear Hormone Receptors in GnRH Neurons in a Male Cichlid Fish. Int J Mol Sci 2020; 21:ijms21082724. [PMID: 32326396 PMCID: PMC7215790 DOI: 10.3390/ijms21082724] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 01/16/2020] [Accepted: 01/17/2020] [Indexed: 12/17/2022] Open
Abstract
Gonadotropin-releasing hormone (GnRH) is essential for the initiation and maintenance of reproductive functions in vertebrates. To date, three distinct paralogue lineages, GnRH1, GnRH2, and GnRH3, have been identified with different functions and regulatory mechanisms. Among them, hypothalamic GnRH1 neurons are classically known as the hypophysiotropic form that is regulated by estrogen feedback. However, the mechanism of action underlying the estrogen-dependent regulation of GnRH1 has been debated, mainly due to the coexpression of low levels of estrogen receptor (ER) genes. In addition, the role of sex steroids in the modulation of GnRH2 and GnRH3 neurons has not been fully elucidated. Using single-cell real-time PCR, we revealed the expression of genes for estrogen, androgen, glucocorticoid, thyroid, and xenobiotic receptors in GnRH1, GnRH2, and GnRH3 neurons in the male Nile tilapia Oreochromis niloticus. We further quantified expression levels of estrogen receptor genes (ERα, ERβ, and ERγ) in three GnRH neuron types in male tilapia of two different social statuses (dominant and subordinate) at the single cell level. In dominant males, GnRH1 mRNA levels were positively proportional to ERγ mRNA levels, while in subordinate males, GnRH2 mRNA levels were positively proportional to ERβ mRNA levels. These results indicate that variations in the expression of nuclear receptors (and possibly steroid sensitivities) among individual GnRH cells may facilitate different physiological processes, such as the promotion of reproductive activities through GnRH1 neurons, and the inhibition of feeding and sexual behaviors through GnRH2 neurons.
Collapse
|
17
|
Bafor EE, Ukpebor F, Elvis-Offiah U, Uchendu A, Omoruyi O, Omogiade GU. Justicia flava Leaves Exert Mild Estrogenic Activity in Mouse Models of Uterotrophic and Reproductive Cycle Investigations. J Med Food 2020; 23:395-408. [DOI: 10.1089/jmf.2019.0155] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Enitome E. Bafor
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Benin, Benin City, Edo State, Nigeria
| | - Faith Ukpebor
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Benin, Benin City, Edo State, Nigeria
| | - Uloma Elvis-Offiah
- Department of Science and Laboratory Technology, Faculty of Science, University of Benin, Benin City, Edo State, Nigeria
| | - Adaeze Uchendu
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Benin, Benin City, Edo State, Nigeria
| | - Osemelomen Omoruyi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Benin, Benin City, Edo State, Nigeria
| | - Glory Uyi Omogiade
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Benin, Benin City, Edo State, Nigeria
| |
Collapse
|
18
|
Sen A, Hoffmann HM. Role of core circadian clock genes in hormone release and target tissue sensitivity in the reproductive axis. Mol Cell Endocrinol 2020; 501:110655. [PMID: 31756424 PMCID: PMC6962569 DOI: 10.1016/j.mce.2019.110655] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 11/15/2019] [Accepted: 11/16/2019] [Indexed: 12/17/2022]
Abstract
Precise timing in hormone release from the hypothalamus, the pituitary and ovary is critical for fertility. Hormonal release patterns of the reproductive axis are regulated by a feedback loop within the hypothalamic-pituitary-gonadal (HPG) axis. The timing and rhythmicity of hormone release and tissue sensitivity in the HPG axis is regulated by circadian clocks located in the hypothalamus (suprachiasmatic nucleus, kisspeptin and GnRH neurons), the pituitary (gonadotrophs), the ovary (theca and granulosa cells), the testis (Leydig cells), as well as the uterus (endometrium and myometrium). The circadian clocks integrate environmental and physiological signals to produce cell endogenous rhythms generated by a transcriptional-translational feedback loop of transcription factors that are collectively called the "molecular clock". This review specifically focuses on the contribution of molecular clock transcription factors in regulating hormone release patterns in the reproductive axis, with an emphasis on the female reproductive system. Specifically, we discuss the contributions of circadian rhythms in distinct neuronal populations of the female hypothalamus, the molecular clock in the pituitary and its overall impact on female and male fertility.
Collapse
Affiliation(s)
- Aritro Sen
- Department of Animal Science and the Reproductive and Developmental Science Program, Michigan State University, East Lansing, MI, 48824, USA
| | - Hanne M Hoffmann
- Department of Animal Science and the Reproductive and Developmental Science Program, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
19
|
Abstract
Estrogens are critical in driving sex-typical social behaviours that are ethologically relevant in mammals. This is due to both production of local estrogens and signaling by these ligands, particularly in an interconnected set of nuclei called the social behavioural network (SBN). The SBN is a sexually dimorphic network studied predominantly in rodents that is thought to underlie the display of social behaviour in mammals. Signalling by the predominant endogenous estrogen, 17β-estradiol, can be either via the classical genomic or non-classical rapid pathway. In the classical genomic pathway, 17β-estradiol binds the intracellular estrogen receptors (ER) α and β which act as ligand-dependent transcription factors to regulate transcription. In the non-genomic pathway, 17β-estradiol binds a putative plasma membrane ER (mER) such as GPR30/GPER1 to rapidly signal via kinases or calcium flux. Though GPER1's role in sexual dimorphism has been explored to a greater extent in cardiovascular physiology, less is known about its role in the brain. In the last decade, activation of GPER1 has been shown to be important for lordosis and social cognition in females. In this review we will focus on several mechanisms that may contribute to sexually dimorphic behaviors including the colocalization of these estrogen receptors in the SBN, interplay between the signaling pathways activated by these different estrogen receptors, and the role of these receptors in development and the maintenance of the SBN, all of which remain underexplored.
Collapse
|
20
|
Roque C, Mendes-Oliveira J, Duarte-Chendo C, Baltazar G. The role of G protein-coupled estrogen receptor 1 on neurological disorders. Front Neuroendocrinol 2019; 55:100786. [PMID: 31513775 DOI: 10.1016/j.yfrne.2019.100786] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 09/02/2019] [Accepted: 09/07/2019] [Indexed: 02/06/2023]
Abstract
G protein-coupled estrogen receptor 1 (GPER) is a membrane-associated estrogen receptor (ER) associated with rapid estrogen-mediated effects. Over recent years GPER emerged has a potential therapeutic target to induce neuroprotection, avoiding the side effects elicited by the activation of classical ERs. The putative neuroprotection triggered by GPER selective activation was demonstrated in mood disorders, Alzheimer's disease or Parkinson's disease of male and female in vivo rodent models. In others, like ischemic stroke, the results are contradictory and currently there is no consensus on the role played by this receptor. However, it seems clear that sex is a biological variable that may impact the results. The major objective of this review is to provide an overview about the physiological effects of GPER in the brain and its putative contribution in neurodegenerative disorders, discussing the data about the signaling pathways involved, as well as, the diverse effects observed.
Collapse
Affiliation(s)
- C Roque
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - J Mendes-Oliveira
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - C Duarte-Chendo
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - G Baltazar
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal; Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal.
| |
Collapse
|
21
|
Moore AM, Abbott G, Mair J, Prescott M, Campbell RE. Mapping GABA and glutamate inputs to gonadotrophin-releasing hormone neurones in male and female mice. J Neuroendocrinol 2018; 30:e12657. [PMID: 30415474 DOI: 10.1111/jne.12657] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 10/22/2018] [Accepted: 11/05/2018] [Indexed: 11/28/2022]
Abstract
Gonadotrophin-releasing hormone (GnRH) neurone function is dependent upon gonadal steroid hormone feedback, which is communicated in large part through an afferent neuronal network. The classical neurotransmitters GABA and glutamate are important regulators of GnRH neurone activity and are implicated in mediating feedback signals. In the present study, we aimed to determine whether GABAergic or glutamatergic input to GnRH neurones differs between males and females and/or exhibits morphological plasticity in response to steroid hormone feedback in females. Tissue collected from GnRH-green fluorescent protein (GFP) male and female mice in dioestrus underwent immunofluorescence labelling of GFP and either the vesicular GABA transporter (VGAT) or the vesicular glutamate transporter 2 (VGLUT2). No differences in the densities or absolute numbers of VGAT-immunoreactive (-IR) or VGLUT2-IR puncta apposed to GnRH neurones were identified between males and females. The most significant input from either neurotransmitter was to the proximal dendritic region and 80% of VGAT-IR puncta apposed to GnRH neurones co-localised with synaptophysin. Putative inputs were also assessed in ovariectomised (OVX) female mice treated with negative (OVX+E) or positive (OVX+E+E) feedback levels of oestrogen, and OVX+E+E mice were killed during the expected GnRH/luteinising hormone surge. No differences in VGLUT2-IR contacts to GnRH neurones were identified between animals under the negative-feedback influence of oestrogen (OVX+E) or the positive influence of oestrogen (OVX+E+E), regardless of cFos activation status. By contrast, a significant elevation in putative GABAergic inputs to GnRH neurones at the time of the preovulatory surge was found in the cFos-negative subset of GnRH neurones, both at the level of the soma and at the proximal dendrite. Taken together, these data suggest that, although GABAergic and glutamatergic innervation of GnRH neurones is not sexually differentiated, cyclic fluctuations in steroid hormone feedback over the female oestrous cycle result in plastic changes in GABAergic inputs to a subpopulation of GnRH neurones.
Collapse
Affiliation(s)
- Aleisha M Moore
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, Mississippi
| | - Georgina Abbott
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Jonathan Mair
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Melanie Prescott
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Rebecca E Campbell
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
22
|
A review on role of medicinal plants in polycystic ovarian syndrome: Pathophysiology, neuroendocrine signaling, therapeutic status and future prospects. MIDDLE EAST FERTILITY SOCIETY JOURNAL 2018. [DOI: 10.1016/j.mefs.2018.04.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
|
23
|
Ma Y, Zhu M, Miao L, Zhang X, Dong X, Zou X. Mercuric Chloride Induced Ovarian Oxidative Stress by Suppressing Nrf2-Keap1 Signal Pathway and its Downstream Genes in Laying Hens. Biol Trace Elem Res 2018; 185:185-196. [PMID: 29349677 DOI: 10.1007/s12011-018-1244-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 01/09/2018] [Indexed: 12/13/2022]
Abstract
The present study evaluated the effects of mercury chloride (HgCl2) on follicular atresia rate, sex hormone secretion, and ovarian oxidative stress in laying hens. Antioxidant enzyme genes and the nuclear factor erythroid 2-related factor 2 (Nrf2)-Kelch-like ECH-associated protein 1 (Keap1) signal pathway were further studied to uncover the molecular mechanism. A total of 768 40-week-old Hy-Line Brown laying hens were randomly allocated to four treatments with eight pens per treatment and 24 hens of each pen. The birds were fed with four experimental diets containing graded levels of mercury (Hg) at 0.280, 3.325, 9.415, and 27.240 mg/kg, respectively. Results revealed that a positive relationship occurred between the accumulation of Hg in ovary and follicular atresia rate. Progesterone (P4) level significantly decreased in all Hg-treatment groups (P < 0.05), and follicle-stimulating hormone (FSH) and luteinizing hormone (LH) levels were the lowest in the 27.240-mg/kg Hg group. Besides, the activities of catalase (CAT), superoxidative dismutase (SOD), glutathione reductase (GR), and glutathione (GSH) content were significantly decreased in all Hg-treatment groups (P < 0.05). Glutathione peroxidase (GSH-Px) activity significantly decreased, while malondialdehyde (MDA) content sharply increased in the 27.240-mg/kg Hg group (P < 0.05). In addition, there were positive relationships between antioxidant enzyme activities and antioxidant gene expressions or between antioxidant gene expressions and Nrf2 mRNA expression, while negative correlations occurred between Nrf2 and Keap1 at transcription and protein levels. It could be concluded that Hg induced ovarian function disorders and ovarian oxidative stress by means of impairing the Nrf2-Keap1 signal pathway in laying hens.
Collapse
Affiliation(s)
- Yan Ma
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Feed Science Institute, College of Animal Science, Zhejiang University, Hangzhou, 310058, China
| | - Mingkun Zhu
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Feed Science Institute, College of Animal Science, Zhejiang University, Hangzhou, 310058, China
| | - Liping Miao
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Feed Science Institute, College of Animal Science, Zhejiang University, Hangzhou, 310058, China
| | - Xiaoyun Zhang
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Feed Science Institute, College of Animal Science, Zhejiang University, Hangzhou, 310058, China
| | - Xinyang Dong
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Feed Science Institute, College of Animal Science, Zhejiang University, Hangzhou, 310058, China
| | - Xiaoting Zou
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Feed Science Institute, College of Animal Science, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
24
|
Loutchanwoot P, Vortherms T. Effects of puerarin on estrogen-regulated gene expression in gonadotropin-releasing hormone pulse generator of ovariectomized rats. Steroids 2018; 135:54-62. [PMID: 29733861 DOI: 10.1016/j.steroids.2018.05.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 04/28/2018] [Accepted: 05/02/2018] [Indexed: 11/28/2022]
Abstract
Effects of puerarin on the hypothalamic gonadotropin-releasing hormone (GnRH) pulse generator function is investigated, for the first time, in ovariectomized rats at the level of mRNA expression of estrogen-responsive genes, e.g., estrogen receptor (ER), GnRH and its receptor (GnRHR). Rats were treated orally for 90 days either with a soy-free diet containing two different doses of puerarin (low dose of 600 mg/kg and high dose of 3000 mg/kg) or estradiol benzoate (E2B) at either low dose (4.3 mg/kg) or high dose (17.3 mg/kg). Levels of mRNA expression in the medial preoptic area/anterior hypothalamus (MPOA/AH), mediobasal hypothalamus/median eminence (MBH/ME) and adenohypophysis were measured by quantitative TaqMan® real-time RT-PCR. Plasma levels of luteinizing hormone (LH) and prolactin (PRL) were measured by radioimmunoassay. In the MPOA/AH, both puerarin and E2B decreased ERα mRNA levels without any significant changes in ERβ and GnRH mRNA levels. Both puerarin and E2B did not significantly alter the expression levels of ERα, ERβ and GnRHR in the MBH/ME. E2B exerted significant effects on the down-regulation of adenohypophyseal GnRHR mRNA transcripts and serum LH levels. Puerarin did not cause significant changes in pituitary GnRHR mRNA transcripts and serum LH and PRL levels. This is the first study to demonstrate that in ovariectomized rat models of ovarian hormone deprivation, puerarin acted as a weak estrogen-active compound in the hypothalamic GnRH pulse generator through the downregulation of MPOA/AH ERα mRNA expression.
Collapse
Affiliation(s)
- Panida Loutchanwoot
- Department of Biology, Faculty of Science, Mahasarakham University, Khamriang Sub-district, Kantarawichai District, Mahasarakham Province 44150, Thailand.
| | - Tina Vortherms
- Department of Endocrinology, Faculty of Medicine, University Medical Center Göttingen, Robert-Koch-Strasse 40, D-37075 Göttingen, Germany
| |
Collapse
|
25
|
Tonn Eisinger KR, Larson EB, Boulware MI, Thomas MJ, Mermelstein PG. Membrane estrogen receptor signaling impacts the reward circuitry of the female brain to influence motivated behaviors. Steroids 2018; 133:53-59. [PMID: 29195840 PMCID: PMC5864533 DOI: 10.1016/j.steroids.2017.11.013] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 11/20/2017] [Accepted: 11/23/2017] [Indexed: 12/12/2022]
Abstract
Within the adult female, estrogen signaling is well-described as an integral component of the physiologically significant hypothalamic-pituitary-gonadal axis. In rodents, the timing of ovulation is intrinsically entwined with the display of sexual receptivity. For decades, the importance of estradiol activating intracellular estrogen receptors within the hypothalamus and midbrain/spinal cord lordosis circuits has been appreciated. These signaling pathways primarily account for the ability of the female to reproduce. Yet, often overlooked is that the desire to reproduce is also tightly regulated by estrogen receptor signaling. This lack of emphasis can be attributed to an absence of nuclear estrogen receptors in brain regions associated with reward, such as the nucleus accumbens, which are associated with motivated behaviors. This review outlines how membrane-localized estrogen receptors affect metabotropic glutamate receptor signaling within the rodent nucleus accumbens. In addition, we discuss how, as estrogens drive increased motivation for reproduction, they also produce the untoward side effect of heightening female vulnerability to drug addiction.
Collapse
Affiliation(s)
- Katherine R Tonn Eisinger
- Department of Neuroscience and Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Erin B Larson
- Department of Neuroscience and Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Marissa I Boulware
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Mark J Thomas
- Department of Neuroscience and Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Paul G Mermelstein
- Department of Neuroscience and Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
26
|
Prepubertal Development of GABAergic Transmission to Gonadotropin-Releasing Hormone (GnRH) Neurons and Postsynaptic Response Are Altered by Prenatal Androgenization. J Neurosci 2018; 38:2283-2293. [PMID: 29374136 DOI: 10.1523/jneurosci.2304-17.2018] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 12/29/2017] [Accepted: 01/20/2018] [Indexed: 11/21/2022] Open
Abstract
Gonadotropin-releasing hormone (GnRH) neurons regulate reproduction through pulsatile GnRH release. Women with polycystic ovary syndrome (PCOS) have persistently elevated luteinizing hormone release frequency, reflecting GnRH release; this exacerbates hyperandrogenemia and disrupted reproductive cycles that are characteristic of this disorder. Clinical evidence suggests that neuroendocrine features of PCOS may manifest peripubertally. Adult mice prenatally exposed to androgens (PNA) mimic several reproductive features of PCOS. GnRH neurons from these mice have increased firing activity and receive increased GABAergic transmission, which is excitatory. When changes emerge during development is unknown. To study the typical postnatal development of GABAergic transmission and the effects of PNA treatment and sex, whole-cell voltage-clamp recordings were made of GABAergic postsynaptic currents (PSCs) in GnRH neurons in brain slices from prepubertal through adult control and PNA female and male mice. GABAergic transmission was present by 1 week of age in females and males and increased in frequency, reaching adult levels at 3 and 4 weeks, respectively. GABAergic PSC frequency was elevated in 3-week-old PNA versus control females. PSC frequency in both controls and PNA mice was activity independent, suggesting that PNA induces changes in synapse organization. PNA also alters the functional response of GnRH neurons to GABA. GABA induced firing in fewer neurons from 3-week-old PNA than control females; membrane potential depolarization induced by GABA was also reduced in cells from PNA mice at this age. PNA thus induces changes during development in the presynaptic organization of the GABAergic network afferent to GnRH neurons as well as the postsynaptic GnRH neuron response, both of which may contribute to adult reproductive dysfunction.SIGNIFICANCE STATEMENT The central neuronal network that regulates reproduction is overactive in polycystic ovary syndrome (PCOS), a leading cause of infertility. Recent evidence of neuroendocrine dysfunction in midpubertal girls suggests that the pathophysiological mechanisms underlying PCOS may arise before pubertal maturation. Prenatal exposure to androgens (PNA) in mice mimics several neuroendocrine features of PCOS. GABAergic transmission to gonadotropin-releasing hormone (GnRH) neurons is important for reproduction and is increased in adult PNA mice. The typical development of this network and when changes with PNA and sex arise relative to puberty are unknown. These studies provide evidence that PNA alters prepubertal development of the GABAergic network afferent to GnRH neurons, including both the presynaptic organization and postsynaptic response. These changes may contribute to reproductive dysfunction in adults.
Collapse
|
27
|
Abstract
Reproductive behavior is the behavior related to the production of offspring and includes all aspects from the establishment of mating systems, courtship, sexual behavior, and parturition to the care of young. In this chapter, I outline the hormonal regulation of the estrous cycle, followed by a description of the neural regulation of female sexual behavior. Ovarian hormones play an important role in the induction of ovulation and behavioral estrus, in which they interact closely with several neurotransmitters and neuropeptides to induce sexual behavior. This chapter discusses the latest research on the role of estrogen, progesterone, serotonin, dopamine, noradrenaline, oxytocin, and GABA in female mating behavior. In addition, the most relevant brain areas, such as the preoptic area and the ventromedial nucleus of the hypothalamus, in which these regulations take place, are discussed.
Collapse
Affiliation(s)
- Eelke M S Snoeren
- Department of Psychology, UiT the Arctic University of Norway, Tromsø, Norway.
| |
Collapse
|
28
|
Yang J, Hu S, Rao M, Hu L, Lei H, Wu Y, Wang Y, Ke D, Xia W, Zhu CH. Copper nanoparticle-induced ovarian injury, follicular atresia, apoptosis, and gene expression alterations in female rats. Int J Nanomedicine 2017; 12:5959-5971. [PMID: 28860760 PMCID: PMC5571856 DOI: 10.2147/ijn.s139215] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Numerous studies have reported the accumulation of copper nanoparticles (Cu NPs) in organs and the corresponding damage, although whether Cu NPs can be translocated to the ovaries and their ovarian toxicity are still unknown. In this study, three groups of female rats were injected with 3.12, 6.25, or 12.5 mg/kg Cu NPs for 14 consecutive days. The pathological changes, hormone levels, apoptosis and apoptotic proteins, oxidative stress, and gene expression characteristics in the ovaries were then investigated. The results demonstrated that the Cu NPs exhibited obvious accumulation in the rat ovaries, leading to ovarian injury, an imbalance of sex hormones, and ovarian cell apoptosis. Cu NP exposure activated caspase 3, caspase 8, caspase 9, and tBid, decreased the protein levels of Bcl-2, increased the expression levels of the proteins Bax and cytochrome c, and promoted malondialdehyde (MDA) accumulation and superoxide dismutase (SOD) reduction. Furthermore, gene microarray analysis showed that Cu NPs (12.5 mg/kg/d) caused 321 differentially expressed genes. Of these, 180 and 141 genes were upregulated and downregulated, respectively. Hsd17b1, Hsd3b1, Hsd3b6, and Hsd3b were involved in steroid and hormone metabolism, whereas Mt3 and Cebpb were associated with apoptosis. Overall, these findings provide strong evidence that Cu NPs trigger both intrinsic and extrinsic apoptotic pathways and regulate key ovarian genes in oxidative stress-mediated ovarian dysfunction.
Collapse
Affiliation(s)
- Jing Yang
- Family Planning Research Institute, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei
| | - Shifu Hu
- Family Planning Research Institute, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei
| | - Meng Rao
- Family Planning Research Institute, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei
| | - Lixia Hu
- Department of Histology and Embryology, Preclinical Medicine College, Xinxiang Medical University, Henan Province, Xinxiang
| | - Hui Lei
- Family Planning Research Institute, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei
| | - Yanqing Wu
- Family Planning Research Institute, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei
| | - Yingying Wang
- Family Planning Research Institute, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei
| | - Dandan Ke
- Family Planning Research Institute, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei
| | - Wei Xia
- Family Planning Research Institute, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei.,Reproductive Medicine Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Chang-Hong Zhu
- Family Planning Research Institute, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei.,Reproductive Medicine Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| |
Collapse
|
29
|
Xiang W, Zhang B, Lv F, Feng G, Chen L, Yang F, Zhang K, Cao C, Wang P, Chu M. The potential regulatory mechanisms of the gonadotropin-releasing hormone in gonadotropin transcriptions identified with bioinformatics analyses. Reprod Biol Endocrinol 2017; 15:46. [PMID: 28623929 PMCID: PMC5474292 DOI: 10.1186/s12958-017-0264-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 06/08/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The regulation of gonadotropin synthesis and release by gonadotropin-releasing hormone (GnRH) plays an essential role in the neuroendocrine control of reproduction. However, the mechanisms underlying gonadotropin regulation by GnRH pulse frequency and amplitude are still ambiguous. This study aimed to explore the molecular mechanisms and biological pathways associated with gonadotropin synthesis by GnRH pulse frequencies and amplitudes. METHODS Using GSE63251 datasets downloaded from the Gene Expression Omnibus (GEO), differentially expressed genes (DEGs) were screened by comparing the RNA expression from the GnRH pulse group, the GnRH tonic group and the control group. Pathway enrichment analyses of DEGs was performed, followed by protein-protein interaction (PPI) network construction. Furthermore, sub-network modules were constructed by ClusterONE and GO function and pathways analysed by DAVID. In addition, the relationship between the metabolic pathways and the GnRH pathway was verified in vitro. RESULTS In total, 531 common DEGs were identified in GnRH groups, including 290 up-regulated and 241 down-regulated genes. DEGs predominantly enriched in 16 Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, including 11 up-regulated pathways (signallingsignallingmetabolic pathways, signallingand GnRH signalling pathway) and 5 down-regulated pathways (type II diabetes mellitus). Moreover, FBJ osteosarcoma oncogene (FOS) and jun proto-oncogene (JUN) had higher connectivity degrees in the PPI network. Three modules in the PPI were identified with ClusterONE. The genes in module 1 were significantly enriched in five pathways, including signallingthe insulin resistance and GnRH signalling pathway. The genes in modules 2 and 3 were mainly enriched in metabolic pathways and steroid hormone biosynthesis, respectively. Finally, knockdown leptin receptor (LEPR) and insulin receptor (INSR) reversed the GnRH-modulated metabolic related-gene expression. CONCLUSIONS The present study revealed the involvement of GnRH in the regulation of gonadotropin biosynthesis and metabolism in the maintenance of reproduction, achieved by bioinformatics analyses. This, indicates that the GnRH signalling pathway played a central linkings role in reproductive function and metabolic balance. In addition, the present study identified the difference response between GnRH pulse and GnRH tone, indicated that abnormal GnRH pulse and amplitude may cause disease, which may provide an improved understanding of the GnRH pathway and a new insight for disease diagnosis and treatment.
Collapse
Affiliation(s)
- Wei Xiang
- College of Bioengineering, Chongqing University, Chongqing, 400030 China
| | - Baoyun Zhang
- College of Bioengineering, Chongqing University, Chongqing, 400030 China
| | - Fenglin Lv
- College of Bioengineering, Chongqing University, Chongqing, 400030 China
| | - Guangde Feng
- Sichuan TQLS Animal Husbandry Science and Technology Co.,LTD, City, Mianyang, Sichuan 621000 China
| | - Long Chen
- College of Bioengineering, Chongqing University, Chongqing, 400030 China
| | - Fang Yang
- College of Bioengineering, Chongqing University, Chongqing, 400030 China
| | - Ke Zhang
- College of Bioengineering, Chongqing University, Chongqing, 400030 China
| | - Chunyu Cao
- College of Bioengineering, Chongqing University, Chongqing, 400030 China
| | - Pingqing Wang
- College of Bioengineering, Chongqing University, Chongqing, 400030 China
| | - Mingxing Chu
- Key Laboratory of Farm Animal Genetic Resources and Germplasm Innovation of Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193 China
| |
Collapse
|
30
|
Sena GC, Freitas-Lima LC, Merlo E, Podratz PL, de Araújo JF, Brandão PA, Carneiro MT, Zicker MC, Ferreira AV, Takiya CM, de Lemos Barbosa CM, Morales MM, Santos-Silva AP, Miranda-Alves L, Silva IV, Graceli JB. Environmental obesogen tributyltin chloride leads to abnormal hypothalamic-pituitary-gonadal axis function by disruption in kisspeptin/leptin signaling in female rats. Toxicol Appl Pharmacol 2017; 319:22-38. [DOI: 10.1016/j.taap.2017.01.021] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 01/27/2017] [Accepted: 01/30/2017] [Indexed: 12/15/2022]
|
31
|
Uenoyama Y, Pheng V, Tsukamura H, Maeda KI. The roles of kisspeptin revisited: inside and outside the hypothalamus. J Reprod Dev 2016; 62:537-545. [PMID: 27478063 PMCID: PMC5177970 DOI: 10.1262/jrd.2016-083] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Kisspeptin, encoded by KISS1/Kiss1 gene, is now considered a master regulator of reproductive functions in mammals owing to
its involvement in the direct activation of gonadotropin-releasing hormone (GnRH) neurons after binding to its cognate receptor, GPR54. Ever since the discovery
of kisspeptin, intensive studies on hypothalamic expression of KISS1/Kiss1 and on physiological roles of hypothalamic
kisspeptin neurons have provided clues as to how the brain controls sexual maturation at the onset of puberty and subsequent reproductive performance in
mammals. Additionally, emerging evidence indicates the potential involvement of extra-hypothalamic kisspeptin in reproductive functions. Here, we summarize data
regarding kisspeptin inside and outside the hypothalamus and revisit the physiological roles of central and peripheral kisspeptins in the reproductive functions
of mammals.
Collapse
Affiliation(s)
- Yoshihisa Uenoyama
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | | | | | | |
Collapse
|
32
|
Sex differences in the brain–an interplay of sex steroid hormones and sex chromosomes. Clin Sci (Lond) 2016; 130:1481-97. [DOI: 10.1042/cs20160299] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 05/17/2016] [Indexed: 12/12/2022]
Abstract
Although considerable progress has been made in our understanding of brain function, many questions remain unanswered. The ultimate goal of studying the brain is to understand the connection between brain structure and function and behavioural outcomes. Since sex differences in brain morphology were first observed, subsequent studies suggest different functional organization of the male and female brains in humans. Sex and gender have been identified as being a significant factor in understanding human physiology, health and disease, and the biological differences between the sexes is not limited to the gonads and secondary sexual characteristics, but also affects the structure and, more crucially, the function of the brain and other organs. Significant variability in brain structures between individuals, in addition to between the sexes, is factor that complicates the study of sex differences in the brain. In this review, we explore the current understanding of sex differences in the brain, mostly focusing on preclinical animal studies.
Collapse
|
33
|
Bálint F, Liposits Z, Farkas I. Estrogen Receptor Beta and 2-arachidonoylglycerol Mediate the Suppressive Effects of Estradiol on Frequency of Postsynaptic Currents in Gonadotropin-Releasing Hormone Neurons of Metestrous Mice: An Acute Slice Electrophysiological Study. Front Cell Neurosci 2016; 10:77. [PMID: 27065803 PMCID: PMC4809870 DOI: 10.3389/fncel.2016.00077] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 03/11/2016] [Indexed: 11/25/2022] Open
Abstract
Gonadotropin-releasing hormone (GnRH) neurons are controlled by 17β-estradiol (E2) contributing to the steroid feedback regulation of the reproductive axis. In rodents, E2 exerts a negative feedback effect upon GnRH neurons throughout the estrus-diestrus phase of the ovarian cycle. The present study was undertaken to reveal the role of estrogen receptor subtypes in the mediation of the E2 signal and elucidate the downstream molecular machinery of suppression. The effect of E2 administration at low physiological concentration (10 pM) on GnRH neurons in acute brain slices obtained from metestrous GnRH-green fluorescent protein (GFP) mice was studied under paradigms of blocking or activating estrogen receptor subtypes and interfering with retrograde 2-arachidonoylglycerol (2-AG) signaling. Whole-cell patch clamp recordings revealed that E2 significantly diminished the frequency of spontaneous postsynaptic currents (sPSCs) in GnRH neurons (49.62 ± 7.6%) which effect was abolished by application of the estrogen receptor (ER) α/β blocker Faslodex (1 μM). Pretreatment of the brain slices with cannabinoid receptor type 1 (CB1) inverse agonist AM251 (1 μM) and intracellularly applied endocannabinoid synthesis blocker THL (10 μM) significantly attenuated the effect of E2 on the sPSCs. E2 remained effective in the presence of tetrodotoxin (TTX) indicating a direct action of E2 on GnRH cells. The ERβ specific agonist DPN (10 pM) also significantly decreased the frequency of miniature postsynaptic currents (mPSCs) in GnRH neurons. In addition, the suppressive effect of E2 was completely blocked by the selective ERβ antagonist PHTPP (1 μM) indicating that ERβ is required for the observed rapid effect of the E2. In contrast, the ERα agonist PPT (10 pM) or the membrane-associated G protein-coupled estrogen receptor (GPR30) agonist G1 (10 pM) had no significant effect on the frequency of mPSCs in these neurons. AM251 and tetrahydrolipstatin (THL) significantly abolished the effect of E2 whereas AM251 eliminated the action of DPN on the mPSCs. These data suggest the involvement of the retrograde endocannabinoid mechanism in the rapid direct effect of E2. These results collectively indicate that estrogen receptor beta and 2-AG/CB1 signaling mechanisms are coupled and play an important role in the mediation of the negative estradiol feedback on GnRH neurons in acute slice preparation obtained from intact, metestrous mice.
Collapse
Affiliation(s)
- Flóra Bálint
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of SciencesBudapest, Hungary; Roska Tamás Doctoral School of Sciences and Technology, Faculty of Information Technology and Bionics, Pázmány Péter Catholic UniversityBudapest, Hungary
| | - Zsolt Liposits
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of SciencesBudapest, Hungary; Department of Neuroscience, Faculty of Information Technology and Bionics, Pázmány Péter Catholic UniversityBudapest, Hungary
| | - Imre Farkas
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences Budapest, Hungary
| |
Collapse
|
34
|
Expression of ESR1 in Glutamatergic and GABAergic Neurons Is Essential for Normal Puberty Onset, Estrogen Feedback, and Fertility in Female Mice. J Neurosci 2016; 35:14533-43. [PMID: 26511244 DOI: 10.1523/jneurosci.1776-15.2015] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Circulating estradiol exerts a profound influence on the activity of the gonadotropin-releasing hormone (GnRH) neuronal network controlling fertility. Using genetic strategies enabling neuron-specific deletion of estrogen receptor α (Esr1), we examine here whether estradiol-modulated GABA and glutamate transmission are critical for the functioning of the GnRH neuron network in the female mouse. Using Vgat- and Vglut2-ires-Cre knock-in mice and ESR1 immunohistochemistry, we demonstrate that subpopulations of GABA and glutamate neurons throughout the limbic forebrain express ESR1, with ESR1-GABAergic neurons being more widespread and numerous than ESR1-glutamatergic neurons. We crossed Vgat- and Vglut2-ires-Cre mice with an Esr1(lox/lox) line to generate animals with GABA-neuron-specific or glutamate-neuron-specific deletion of Esr1. Vgat-ires-Cre;Esr1(lox/lox) mice were infertile, with abnormal estrous cycles, and exhibited a complete failure of the estrogen positive feedback mechanism responsible for the preovulatory GnRH surge. However, puberty onset and estrogen negative feedback were normal. Vglut2-ires-Cre;Esr1(lox/lox) mice were also infertile but displayed a wider range of deficits, including advanced puberty onset, abnormal negative feedback, and abolished positive feedback. Whereas <25% of preoptic kisspeptin neurons expressed Cre in Vgat- and Vglut2-ires-Cre lines, ∼70% of arcuate kisspeptin neurons were targeted in Vglut2-ires-Cre;Esr1(lox/lox) mice, possibly contributing to their advanced puberty phenotype. These observations show that, unexpectedly, ESR1-GABA neurons are only essential for the positive feedback mechanism. In contrast, we reveal the key importance of ESR1 in glutamatergic neurons for multiple estrogen feedback loops within the GnRH neuronal network required for fertility in the female mouse.
Collapse
|
35
|
Ubuka T, Son YL, Tsutsui K. Molecular, cellular, morphological, physiological and behavioral aspects of gonadotropin-inhibitory hormone. Gen Comp Endocrinol 2016; 227:27-50. [PMID: 26409890 DOI: 10.1016/j.ygcen.2015.09.009] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 09/09/2015] [Accepted: 09/11/2015] [Indexed: 12/15/2022]
Abstract
Gonadotropin-inhibitory hormone (GnIH) is a hypothalamic neuropeptide that was isolated from the brains of Japanese quail in 2000, which inhibited luteinizing hormone release from the anterior pituitary gland. Here, we summarize the following fifteen years of researches that investigated on the mechanism of GnIH actions at molecular, cellular, morphological, physiological, and behavioral levels. The unique molecular structure of GnIH peptide is in its LPXRFamide (X=L or Q) motif at its C-terminal. The primary receptor for GnIH is GPR147. The cell signaling pathway triggered by GnIH is initiated by inhibiting adenylate cyclase and decreasing cAMP production in the target cell. GnIH neurons regulate not only gonadotropin synthesis and release in the pituitary, but also regulate various neurons in the brain, such as GnRH1, GnRH2, dopamine, POMC, NPY, orexin, MCH, CRH, oxytocin, and kisspeptin neurons. GnIH and GPR147 are also expressed in gonads and they may regulate steroidogenesis and germ cell maturation in an autocrine/paracrine manner. GnIH regulates reproductive development and activity. In female mammals, GnIH may regulate estrous or menstrual cycle. GnIH is also involved in the regulation of seasonal reproduction, but GnIH may finely tune reproductive activities in the breeding seasons. It is involved in stress responses not only in the brain but also in gonads. GnIH may inhibit male socio-sexual behavior by stimulating the activity of cytochrome P450 aromatase in the brain and stimulates feeding behavior by modulating the activities of hypothalamic and central amygdala neurons.
Collapse
Affiliation(s)
- Takayoshi Ubuka
- Department of Biology, Waseda University, 2-2 Wakamatsu-cho, Shinjuku, Tokyo 162-8480, Japan; Brain Research Institute Monash Sunway (BRIMS) of the Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Petaling Jaya 46150, Malaysia.
| | - You Lee Son
- Department of Biology, Waseda University, 2-2 Wakamatsu-cho, Shinjuku, Tokyo 162-8480, Japan
| | - Kazuyoshi Tsutsui
- Department of Biology, Waseda University, 2-2 Wakamatsu-cho, Shinjuku, Tokyo 162-8480, Japan.
| |
Collapse
|
36
|
Russo KA, La JL, Stephens SBZ, Poling MC, Padgaonkar NA, Jennings KJ, Piekarski DJ, Kauffman AS, Kriegsfeld LJ. Circadian Control of the Female Reproductive Axis Through Gated Responsiveness of the RFRP-3 System to VIP Signaling. Endocrinology 2015; 156:2608-18. [PMID: 25872006 PMCID: PMC4475714 DOI: 10.1210/en.2014-1762] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Throughout most of the ovulatory cycle, estrogen negative feedback restrains the GnRH neuronal system. Just before ovulation, however, estrogen negative feedback is removed to permit stimulation of the preovulatory GnRH/LH surge (positive feedback) by the circadian clock in the suprachiasmatic nucleus (SCN). The mammalian ortholog of avian gonadotropin-inhibitory hormone, RFamide-related peptide 3 (RFRP-3), participates in the circadian-timed removal of estrogen negative feedback to permit the LH surge. The present study examined the specific neurochemical means by which the SCN controls RFRP-3 activity and explored whether the RFRP-3 system exhibits time-dependent responsiveness to SCN signaling to precisely time the LH surge. We found that RFRP-3 cells in female Syrian hamsters (Mesocricetus auratus) receive close appositions from SCN-derived vasopressin-ergic and vasoactive intestinal peptide (VIP)-ergic terminal fibers. Central VIP administration markedly suppressed RFRP-3 cellular activity in the evening, but not the morning, relative to saline controls, whereas vasopressin was without effect at either time point. Double-label in situ hybridization for Rfrp-3 and the VIP receptors VPAC1 and VPAC2 revealed that the majority of RFRP-3 cells do not coexpress either receptor in Syrian hamsters or mice, suggesting that SCN VIP-ergic signaling inhibits RFRP-3 cells indirectly. The timing of this VIP-mediated disinhibition is further coordinated via temporally gated responsiveness of RFRP-3 cells to circadian signaling. Together, these findings reveal a novel circadian hierarchy of control coordinating the preovulatory LH surge and ovulation.
Collapse
Affiliation(s)
- Kimberly A Russo
- Department of Psychology (K.A.R., J.L.L., N.A.P., K.J.J., D.J.P., L.J.K.) and The Helen Wills Neuroscience Institute (L.J.K.), University of California, Berkeley, Berkeley, California 94720; and Department of Reproductive Medicine (S.B.Z.S., M.C.P., A.S.K.), University of California, San Diego, La Jolla, California 92093
| | - Janet L La
- Department of Psychology (K.A.R., J.L.L., N.A.P., K.J.J., D.J.P., L.J.K.) and The Helen Wills Neuroscience Institute (L.J.K.), University of California, Berkeley, Berkeley, California 94720; and Department of Reproductive Medicine (S.B.Z.S., M.C.P., A.S.K.), University of California, San Diego, La Jolla, California 92093
| | - Shannon B Z Stephens
- Department of Psychology (K.A.R., J.L.L., N.A.P., K.J.J., D.J.P., L.J.K.) and The Helen Wills Neuroscience Institute (L.J.K.), University of California, Berkeley, Berkeley, California 94720; and Department of Reproductive Medicine (S.B.Z.S., M.C.P., A.S.K.), University of California, San Diego, La Jolla, California 92093
| | - Matthew C Poling
- Department of Psychology (K.A.R., J.L.L., N.A.P., K.J.J., D.J.P., L.J.K.) and The Helen Wills Neuroscience Institute (L.J.K.), University of California, Berkeley, Berkeley, California 94720; and Department of Reproductive Medicine (S.B.Z.S., M.C.P., A.S.K.), University of California, San Diego, La Jolla, California 92093
| | - Namita A Padgaonkar
- Department of Psychology (K.A.R., J.L.L., N.A.P., K.J.J., D.J.P., L.J.K.) and The Helen Wills Neuroscience Institute (L.J.K.), University of California, Berkeley, Berkeley, California 94720; and Department of Reproductive Medicine (S.B.Z.S., M.C.P., A.S.K.), University of California, San Diego, La Jolla, California 92093
| | - Kimberly J Jennings
- Department of Psychology (K.A.R., J.L.L., N.A.P., K.J.J., D.J.P., L.J.K.) and The Helen Wills Neuroscience Institute (L.J.K.), University of California, Berkeley, Berkeley, California 94720; and Department of Reproductive Medicine (S.B.Z.S., M.C.P., A.S.K.), University of California, San Diego, La Jolla, California 92093
| | - David J Piekarski
- Department of Psychology (K.A.R., J.L.L., N.A.P., K.J.J., D.J.P., L.J.K.) and The Helen Wills Neuroscience Institute (L.J.K.), University of California, Berkeley, Berkeley, California 94720; and Department of Reproductive Medicine (S.B.Z.S., M.C.P., A.S.K.), University of California, San Diego, La Jolla, California 92093
| | - Alexander S Kauffman
- Department of Psychology (K.A.R., J.L.L., N.A.P., K.J.J., D.J.P., L.J.K.) and The Helen Wills Neuroscience Institute (L.J.K.), University of California, Berkeley, Berkeley, California 94720; and Department of Reproductive Medicine (S.B.Z.S., M.C.P., A.S.K.), University of California, San Diego, La Jolla, California 92093
| | - Lance J Kriegsfeld
- Department of Psychology (K.A.R., J.L.L., N.A.P., K.J.J., D.J.P., L.J.K.) and The Helen Wills Neuroscience Institute (L.J.K.), University of California, Berkeley, Berkeley, California 94720; and Department of Reproductive Medicine (S.B.Z.S., M.C.P., A.S.K.), University of California, San Diego, La Jolla, California 92093
| |
Collapse
|
37
|
Hala D, Petersen LH, Martinović D, Huggett DB. In Silico analysis of perturbed steroidogenesis and gonad growth in fathead minnows (P. promelas) exposed to 17α-ethynylestradiol. Syst Biol Reprod Med 2015; 61:122-38. [PMID: 25910217 DOI: 10.3109/19396368.2015.1035817] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The multi-factorial nature of adverse reproductive effects mediated by endocrine disrupting compounds (or EDCs) makes understanding the mechanistic basis of reproductive dysfunction a highly pertinent area of research. As a consequence, a main motivator for continued research is to integrate 'multi-leveled' complexity (i.e., from genes to phenotype) using mathematical methods capable of encapsulating properties of physiological relevance. In this study, an in silico stoichiometric model of piscine steroidogenesis was augmented with a 'biomass' reaction associating the underlying stoichiometry of steroidogenesis with a reaction representative of gonad growth. The ability of the in silico model to predict perturbed steroidogenesis and subsequent effects on gonad growth was tested by exposing reproductively active male and female fathead minnows (Pimephales promelas) to 88 ng/L of the synthetic estrogen, 17α-ethynylestradiol (EE2). The in silico model was parameterized (or constrained) with experimentally quantified concentrations of selected steroid hormones (using mass spectrometry) and fold changes in gene expression (using RT-qPCR) for selected steroidogenic enzyme genes, in gonads of male and female fish. Once constrained, the optimization framework of flux balance analysis (FBA) was used to calculate an optimal flux through the biomass reaction (analogous to gonad growth) and associated steroidogenic flux distributions required to generate biomass. FBA successfully predicted effects of EE2 exposure on fathead minnow gonad growth (%gonadosomatic index or %GSI) and perturbed production of steroid hormones. Specifically, FBA accurately predicted no effects of exposure on male %GSI and a significant reduction for female %GSI. Furthermore, in silico simulations accurately identified disrupted reaction fluxes catalyzing productions of androgens (in male fish) and progestogens (in female fish), an observation which agreed with in vivo experimentation. The analyses presented is the first-ever to successfully associate underlying flux properties of the steroidogenic network with gonad growth in fish, an approach which can incorporate in silico predictions with toxicological risk assessments.
Collapse
Affiliation(s)
- David Hala
- Department of Biology, University of North Texas , Denton, TX , USA
| | | | | | | |
Collapse
|
38
|
Simonneaux V, Bahougne T. A Multi-Oscillatory Circadian System Times Female Reproduction. Front Endocrinol (Lausanne) 2015; 6:157. [PMID: 26539161 PMCID: PMC4611855 DOI: 10.3389/fendo.2015.00157] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 09/21/2015] [Indexed: 01/14/2023] Open
Abstract
Rhythms in female reproduction are critical to insure that timing of ovulation coincides with oocyte maturation and optimal sexual arousal. This fine tuning of female reproduction involves both the estradiol feedback as an indicator of oocyte maturation, and the master circadian clock of the suprachiasmatic nuclei (SCN) as an indicator of the time of the day. Herein, we are providing an overview of the state of knowledge regarding the differential inhibitory and stimulatory effects of estradiol at different stages of the reproductive axis, and the mechanisms through which the two main neurotransmitters of the SCN, arginine vasopressin, and vasoactive intestinal peptide, convey daily time cues to the reproductive axis. In addition, we will report the most recent findings on the putative functions of peripheral clocks located throughout the reproductive axis [kisspeptin (Kp) neurons, gonadotropin-releasing hormone neurons, gonadotropic cells, the ovary, and the uterus]. This review will point to the critical position of the Kp neurons of the anteroventral periventricular nucleus, which integrate both the stimulatory estradiol signal, and the daily arginine vasopressinergic signal, while displaying a circadian clock. Finally, given the critical role of the light/dark cycle in the synchronization of female reproduction, we will discuss the impact of circadian disruptions observed during shift-work conditions on female reproductive performance and fertility in both animal model and humans.
Collapse
Affiliation(s)
- Valérie Simonneaux
- Institut des Neurosciences Cellulaires et Intégratives, CNRS (UPR 3212), Strasbourg, France
- *Correspondence: Valérie Simonneaux, Institut des Neurosciences Cellulaires et Intégratives, CNRS (UPR 3212), 5 rue Blaise Pascal, Strasbourg 67084, France,
| | - Thibault Bahougne
- Institut des Neurosciences Cellulaires et Intégratives, CNRS (UPR 3212), Strasbourg, France
- Service d’Endocrinologie et Diabète, Hôpital Civil, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| |
Collapse
|
39
|
Eghlidi DH, Urbanski HF. Effects of Age and Estradiol on Gene Expression in the Rhesus Macaque Hypothalamus. Neuroendocrinology 2015; 101:236-45. [PMID: 25765287 PMCID: PMC4475460 DOI: 10.1159/000381063] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 02/18/2015] [Indexed: 11/19/2022]
Abstract
BACKGROUND The hypothalamus plays a key role in mediating the effects of estrogen on many physiological functions, including reproduction, metabolism, and thermoregulation. We have previously observed marked estrogen-dependent gene expression changes within the hypothalamus of rhesus macaques during aging, especially in the KNDy neurons of the arcuate-median eminence (ARC-ME) that produce kisspeptin, neurokinin B, and dynorphin A. Little is known, however, about the mechanisms involved in mediating the feedback from estrogen onto these neurons. METHODS We used quantitative real-time PCR to profile age- and estrogen-dependent gene expression changes in the rhesus macaque hypothalamus. Our focus was on genes that encode steroid receptors (ESR1, ESR2, PGR, and AR) and on enzymes that contribute to the local synthesis of 17β-estradiol (E2; STS, HSD3B1/2, HSD17B5, and CYP19A). In addition, we used RT(2) Profiler™ PCR Arrays to profile a larger set of genes that are integral to hypothalamic function. RESULTS KISS1, KISS1R, TAC3, and NPY2R mRNA levels increased in surgically menopausal (ovariectomized) old females relative to age-matched ovariectomized animals that received E2 hormone therapy. In contrast, PGR, HSD17B, GNRH2, SLC6A3, KISS1, TAC3, and NPY2R mRNA levels increased after E2 supplementation. CONCLUSION The rhesus macaque ARC-ME expresses many genes that are responsive to changes in circulating estrogen levels, even during old age, and these may contribute to causing the normal and pathophysiological changes that occur during menopause.
Collapse
Affiliation(s)
- Dominique H. Eghlidi
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oreg., USA
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, Oreg., USA
| | - Henryk F. Urbanski
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oreg., USA
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, Oreg., USA
- Deptartment of Physiology and Pharmacology, Oregon Health and Science University, Portland, Oreg., USA
| |
Collapse
|
40
|
Zaid SSM, Othman S, Kassim NM. Potential protective effect of Tualang honey on BPA-induced ovarian toxicity in prepubertal rat. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 14:509. [PMID: 25519484 PMCID: PMC4301897 DOI: 10.1186/1472-6882-14-509] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 12/09/2014] [Indexed: 12/23/2022]
Abstract
BACKGROUND To investigate the potential protective effects of Tualang honey against the toxicity effects induced by Bisphenol A (BPA) on pubertal development of ovaries. METHODS This study was conducted on pre-pubertal female Sprague Dawley rats. Animals were divided into four groups (n = 8 in each group). Group I was administered with vehicle 0.2 ml of corn oil (Sigma-Aldrich, USA) using oral gavage daily for six weeks; these animals served as negative control (CO group), Group II was administered with BPA suspended in corn oil at 10 mg/kg body weight and served as positive control (PC group), Group III was administered with 200 mg/kg body weight of Tualang honey 30 min before the administration of BPA at 10 mg/kg (TH group) while Group IV was administered with 200 mg/kg body weight of Tualang honey 30 min before the administration of corn oil (THC group). Body weight of all animals were monitored weekly. RESULTS The BPA-exposed animals exhibited disruption of their estrus cycle, while those animals treated with BPA together with Tualang honey, exhibited an improvement in percentage of normal estrous cycle. Their ovaries had lower numbers of atretic follicles compared to the PC group but higher than the CO group. CONCLUSIONS Tualang honey has a potential role in reducing BPA-induced ovarian toxicity by reducing the morphological abnormalities of the ovarian follicles and improving the normal estrous cycle.
Collapse
|
41
|
Yeo SH, Herbison AE. Estrogen-negative feedback and estrous cyclicity are critically dependent upon estrogen receptor-α expression in the arcuate nucleus of adult female mice. Endocrinology 2014; 155:2986-95. [PMID: 24905671 DOI: 10.1210/en.2014-1128] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The location and characteristics of cells within the brain that suppress GnRH neuron activity to contribute to the estrogen-negative feedback mechanism are poorly understood. Using adeno-associated virus (AAV)-mediated Cre-LoxP recombination in estrogen receptor-α (ERα) floxed mice (ERα(flox/flox)), we aimed to examine the role of ERα-expressing neurons located in the arcuate nucleus (ARN) in the estrogen-negative feedback mechanism. Bilateral injection of AAV-Cre into the ARN of ERα(flox/flox) mice (n = 14) resulted in the time-dependent ablation of up to 99% of ERα-immunoreactive cell numbers throughout the rostrocaudal length of the ARN. These mice were all acyclic by 5 weeks after AAV-Cre injections with most mice in constant estrous. Control wild-type mice injected with AAV-Cre (n = 13) were normal. Body weight was not altered in ERα(flox/flox) mice. After ovariectomy, a significant increment in LH secretion was observed in all genotypes, although its magnitude was reduced in ERα(flox/flox) mice. Acute and chronic estrogen-negative feedback were assessed by administering 17β-estradiol to mice as a bolus (LH measured 3 h later) or SILASTIC brand capsule implant (LH measured 5 d later). This demonstrated that chronic estrogen feedback was absent in ERα(flox/flox) mice, whereas the acute feedback was normal. These results reveal a critical role for ERα-expressing cells within the ARN in both estrous cyclicity and the chronic estrogen negative feedback mechanism in female mice. This suggests that ARN cells provide a key indirect, transsynpatic route through which estradiol suppresses the activity of GnRH neurons.
Collapse
Affiliation(s)
- Shel-Hwa Yeo
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Medical Sciences, Dunedin 9054, New Zealand
| | | |
Collapse
|
42
|
Naugle MM, Nguyen LT, Merceron TK, Filardo E, Janssen WGM, Morrison JH, Rapp PR, Gore AC. G-protein coupled estrogen receptor, estrogen receptor α, and progesterone receptor immunohistochemistry in the hypothalamus of aging female rhesus macaques given long-term estradiol treatment. ACTA ACUST UNITED AC 2014; 321:399-414. [PMID: 24862737 DOI: 10.1002/jez.1871] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 04/28/2014] [Indexed: 12/15/2022]
Abstract
Steroid hormone receptors are widely and heterogeneously expressed in the brain, and are regulated by age and gonadal hormones. Our goal was to quantify effects of aging, long-term estradiol (E2 ) treatment, and their interactions, on expression of G protein-coupled estrogen receptor (GPER), estrogen receptor α (ERα) and progesterone receptor (PR) immunoreactivity in two hypothalamic regions, the arcuate (ARC) and the periventricular area (PERI) of rhesus monkeys as a model of menopause and hormone replacement. Ovariectomized (OVX) rhesus macaques were young (∼ 11 years) or aged (∼ 25 years), given oil (vehicle) or E2 every 3 weeks for 2 years. Immunohistochemistry and stereologic analysis of ERα, PR, and GPER was performed. More effects were detected for GPER than the other two receptors. Specifically, GPER cell density in the ARC and PERI, and the percent of GPER-immunoreactive cells in the PERI, were greater in aged than in young monkeys. In addition, we mapped the qualitative distribution of GPER in the monkey hypothalamus and nearby regions. For ERα, E2 treated monkeys tended to have higher cell density than vehicle monkeys in the ARC. The percent of PR density in the PERI tended to be higher in E2 than vehicle monkeys of both ages. This study shows that the aged hypothalamus maintains expression of hormone receptors with age, and that long-term cyclic E2 treatment has few effects on their expression, although GPER was affected more than ERα or PR. This result is surprising in light of evidence for E2 regulation of the receptors studied here, and differences may be due to the selected regions, long-term nature of E2 treatment, among other possibilities.
Collapse
Affiliation(s)
- Michelle M Naugle
- Institute for Neuroscience, University of Texas at Austin, Austin, Texas
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Cheong RY, Porteous R, Chambon P, Abrahám I, Herbison AE. Effects of neuron-specific estrogen receptor (ER) α and ERβ deletion on the acute estrogen negative feedback mechanism in adult female mice. Endocrinology 2014; 155:1418-27. [PMID: 24476134 DOI: 10.1210/en.2013-1943] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The negative feedback mechanism through which 17β-estradiol (E2) acts to suppress the activity of the GnRH neurons remains unclear. Using inducible and cell-specific genetic mouse models, we examined the estrogen receptor (ER) isoforms expressed by neurons that mediate acute estrogen negative feedback. Adult female mutant mice in which ERα was deleted from all neurons in the neonatal period failed to exhibit estrous cycles or negative feedback. Adult mutant female mice with neonatal neuronal ERβ deletion exhibited normal estrous cycles, but a failure of E2 to suppress LH secretion was seen in ovariectomized mice. Mutant mice with a GnRH neuron-selective deletion of ERβ exhibited normal cycles and negative feedback, suggesting no critical role for ERβ in GnRH neurons in acute negative feedback. To examine the adult roles of neurons expressing ERα, an inducible tamoxifen-based Cre-LoxP approach was used to ablate ERα from neurons that express calmodulin kinase IIα in adults. This resulted in mice with no estrous cycles, a normal increase in LH after ovariectomy, but an inability of E2 to suppress LH secretion. Finally, acute administration of ERα- and ERβ-selective agonists to adult ovariectomized wild-type mice revealed that activation of ERα suppressed LH secretion, whereas ERβ agonists had no effect. This study highlights the differences in adult reproductive phenotypes that result from neonatal vs adult ablation of ERα in the brain. Together, these experiments expand previous global knockout studies by demonstrating that neurons expressing ERα are essential and probably sufficient for the acute estrogen negative feedback mechanism in female mice.
Collapse
Affiliation(s)
- Rachel Y Cheong
- Centre for Neuroendocrinology (R.Y.C., R.P., I.A., A.E.H.), Department of Physiology, University of Otago School of Medical Sciences, Dunedin 9054, New Zealand; and Institut de Génétique et de Biologie Moléculaire et Cellulaire (P.C.), 67400 Illkirch, France
| | | | | | | | | |
Collapse
|
44
|
Antonson P, Matic M, Portwood N, Kuiper RV, Bryzgalova G, Gao H, Windahl SH, Humire P, Ohlsson C, Berggren PO, Gustafsson JÅ, Dahlman-Wright K. aP2-Cre-mediated inactivation of estrogen receptor alpha causes hydrometra. PLoS One 2014; 9:e85581. [PMID: 24416430 PMCID: PMC3885723 DOI: 10.1371/journal.pone.0085581] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 11/29/2013] [Indexed: 01/18/2023] Open
Abstract
In this study we describe the reproductive phenotypes of a novel mouse model in which Cre-mediated deletion of ERα is regulated by the aP2 (fatty acid binding protein 4) promoter. ERα-floxed mice were crossed with transgenic mice expressing Cre-recombinase under the control of the aP2 promoter to generate aP2-Cre/ERα(flox/flox) mice. As expected, ERα mRNA levels were reduced in adipose tissue, but in addition we also detected an 80% reduction of ERα levels in the hypothalamus of aP2-Cre/ERα(flox/flox) mice. Phenotypic analysis revealed that aP2-Cre/ERα(flox/flox) female mice were infertile. In line with this, aP2-Cre/ERα(flox/flox) female mice did not cycle and presented 3.8-fold elevated estrogen levels. That elevated estrogen levels were associated with increased estrogen signaling was evidenced by increased mRNA levels of the estrogen-regulated genes lactoferrin and aquaporin 5 in the uterus. Furthermore, aP2-Cre/ERα(flox/flox) female mice showed an accumulation of intra-uterine fluid, hydrometra, without overt indications for causative anatomical anomalies. However, the vagina and cervix displayed advanced keratosis with abnormal quantities of accumulating squamous epithelial cells suggesting functional obstruction by keratin plugs. Importantly, treatment of aP2-Cre/ERα(flox/flox) mice with the aromatase inhibitor Letrozole caused regression of the hydrometra phenotype linking increased estrogen levels to the observed phenotype. We propose that in aP2-Cre/ERα(flox/flox) mice, increased serum estrogen levels cause over-stimulation in the uterus and genital tracts resulting in hydrometra and vaginal obstruction.
Collapse
Affiliation(s)
- Per Antonson
- Department of Biosciences and Nutrition, Karolinska Institutet, Novum, Huddinge, Sweden
| | - Marko Matic
- Department of Biosciences and Nutrition, Karolinska Institutet, Novum, Huddinge, Sweden
| | - Neil Portwood
- The Rolf Luft Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, Stockholm, Sweden
| | - Raoul V Kuiper
- Karolinska Institute Phenotyping Core Facility, Department of Laboratory Medicine, Karolinska University Hospital, Huddinge, Sweden
| | - Galyna Bryzgalova
- The Rolf Luft Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, Stockholm, Sweden
| | - Hui Gao
- Department of Biosciences and Nutrition, Karolinska Institutet, Novum, Huddinge, Sweden
| | - Sara H Windahl
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Patricia Humire
- Department of Biosciences and Nutrition, Karolinska Institutet, Novum, Huddinge, Sweden
| | - Claes Ohlsson
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Per-Olof Berggren
- The Rolf Luft Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, Stockholm, Sweden
| | - Jan-Åke Gustafsson
- Department of Biosciences and Nutrition, Karolinska Institutet, Novum, Huddinge, Sweden ; Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, Texas, United States of America
| | - Karin Dahlman-Wright
- Department of Biosciences and Nutrition, Karolinska Institutet, Novum, Huddinge, Sweden
| |
Collapse
|
45
|
Loutchanwoot P, Srivilai P, Jarry H. Lack of anti-androgenic effects of equol on reproductive neuroendocrine function in the adult male rat. Horm Behav 2014; 65:22-31. [PMID: 24211351 DOI: 10.1016/j.yhbeh.2013.10.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Revised: 10/29/2013] [Accepted: 10/31/2013] [Indexed: 10/26/2022]
Abstract
Equol (EQ), a metabolite of the soy isoflavone daidzein, has well known estrogenic properties. Data from animal studies suggested that EQ may act also as an anti-androgen. However, data regarding how EQ may affect brain functions like the regulation of neuroendocrine activity and reproductive outcomes in adult male rats are still lacking. We therefore investigated the effects of EQ on sex-steroid regulated gene expression in the brain [medial preoptic area/anterior hypothalamus (MPOA/AH) and medial basal hypothalamus/median eminence (MBH/ME)], pituitary, and prostate as a reference androgen-dependent organ. Furthermore reproductive outcomes were evaluated. The anti-androgen flutamide (FLUT) served as reference compound. Male rats (n=12 per group) were treated by gavage for 5 days with either EQ (100 or 250 mg/kgBW/day), or FLUT 100 mg/kgBW/day. All vehicle- and EQ-treated males showed successful reproductive outcomes, whereas FLUT-exposed males had severe reproductive impairments resulted in infertility. FLUT decreased relative weights of prostate, seminal vesicles and epididymides, and increased serum levels of luteinizing hormone, follicle-stimulating hormone, testosterone and 5α-dihydrotestosterone without altering prolactin levels, whereas EQ exerted opposite effects. Both EQ and FLUT decreased gonadotropin releasing hormone (GnRH) expression in the MPOA/AH. Only FLUT upregulated levels of GnRH receptor expression both in the MBH/ME and pituitary. While EQ downregulated the hypothalamic ERα and ERβ expressions, but FLUT did not. In the prostate, only FLUT upregulated both ERα and AR mRNA expression levels. Taken together, our findings are the first data that EQ did not induce anti-androgenic effects on brain, prostate and male reproductive parameters, however, estrogenic neuroendocrine and reproductive effects of EQ were observed.
Collapse
Affiliation(s)
- Panida Loutchanwoot
- Department of Biology, Faculty of Science, Mahasarakham University, Khamriang Sub-district, Kantarawichai District, Mahasarakham Province 44150, Thailand.
| | - Prayook Srivilai
- Department of Biology, Faculty of Science, Mahasarakham University, Khamriang Sub-district, Kantarawichai District, Mahasarakham Province 44150, Thailand.
| | - Hubertus Jarry
- Department of Endocrinology, University Medical Center Göttingen, Robert-Koch-Strasse 40, D-37075 Göttingen, Germany.
| |
Collapse
|
46
|
Yeo SH, Clarkson J, Herbison AE. Kisspeptin-gpr54 signaling at the GnRH neuron is necessary for negative feedback regulation of luteinizing hormone secretion in female mice. Neuroendocrinology 2014; 100:191-7. [PMID: 25301053 DOI: 10.1159/000368608] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 09/10/2014] [Indexed: 11/19/2022]
Abstract
Kisspeptin-Gpr54 signaling is critical for regulating the activity of gonadotropin-releasing hormone (GnRH) neurons in mammals. Previous studies have shown that the negative feedback mechanism is disrupted in global Gpr54-null mutants. The present investigation aimed to determine (1) if a lack of cyclical estrogen exposure of the GnRH neuronal network in the life-long hypogonadotropic Gpr54-null mice contributed to their failed negative feedback mechanism and (2) the cellular location of disrupted kisspeptin-Gpr54 signaling. Plasma luteinizing hormone (LH) concentrations were determined in individual adult female mice when intact, following ovariectomy (OVX) and in response to an acute injection of 17β-estradiol (E2). Control mice exhibited a characteristic rise in LH after OVX that was suppressed by acute E2. Global Gpr54-null mice failed to exhibit any post-OVX increase in LH or response to E2. Adult female global Gpr54-null mice given a cyclical regimen of estradiol for three cycles prior to OVX also failed to exhibit any post-OVX increase in LH or response to E2. To address whether Gpr54 signaling at the GnRH neuron itself was necessary for the failed response to OVX in global Gpr54-null animals, adult female mice with a GnRH neuron-selective deletion of Gpr54 were examined. These mice also failed to exhibit any post-OVX increase in LH or response to E2. These experiments demonstrate defective negative feedback in global Gpr54-null mice that cannot be attributed to a lack of prior exposure of the GnRH neuronal network to cyclical estradiol. The absence of negative feedback in GnRH neuron-selective Gpr54-null mice demonstrates the necessity of direct kisspeptin signaling at the GnRH neuron for this mechanism to occur.
Collapse
Affiliation(s)
- Shel-Hwa Yeo
- Centre for Neuroendocrinology, Department of Physiology, School of Medical Sciences, University of Otago, Dunedin, New Zealand
| | | | | |
Collapse
|
47
|
Terasaka T, Otsuka F, Tsukamoto N, Nakamura E, Inagaki K, Toma K, Ogura-Ochi K, Glidewell-Kenney C, Lawson MA, Makino H. Mutual interaction of kisspeptin, estrogen and bone morphogenetic protein-4 activity in GnRH regulation by GT1-7 cells. Mol Cell Endocrinol 2013; 381:8-15. [PMID: 23880664 PMCID: PMC4079587 DOI: 10.1016/j.mce.2013.07.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 06/12/2013] [Accepted: 07/12/2013] [Indexed: 11/21/2022]
Abstract
Reproduction is integrated by interaction of neural and hormonal signals converging on hypothalamic neurons for controlling gonadotropin-releasing hormone (GnRH). Kisspeptin, the peptide product of the kiss1 gene and the endogenous agonist for the GRP54 receptor, plays a key role in the regulation of GnRH secretion. In the present study, we investigated the interaction between kisspeptin, estrogen and BMPs in the regulation of GnRH production by using mouse hypothalamic GT1-7 cells. Treatment with kisspeptin increased GnRH mRNA expression and GnRH protein production in a concentration-dependent manner. The expression levels of kiss1 and GPR54 were not changed by kisspeptin stimulation. Kisspeptin induction of GnRH was suppressed by co-treatment with BMPs, with BMP-4 action being the most potent for suppressing the kisspeptin effect. The expression of kisspeptin receptor, GPR54, was suppressed by BMPs, and this effect was reversed in the presence of kisspeptin. It was also revealed that BMP-induced Smad1/5/8 phosphorylation and Id-1 expression were suppressed and inhibitory Smad6/7 was induced by kisspeptin. In addition, estrogen induced GPR54 expression, while kisspeptin increased the expression levels of ERα and ERβ, suggesting that the actions of estrogen and kisspeptin are mutually enhanced in GT1-7 cells. Moreover, kisspeptin stimulated MAPKs and AKT signaling, and ERK signaling was functionally involved in the kisspeptin-induced GnRH expression. BMP-4 was found to suppress kisspeptin-induced GnRH expression by reducing ERK signaling activity. Collectively, the results indicate that the axis of kisspeptin-induced GnRH production is bi-directionally controlled, being augmented by an interaction between ERα/β and GPR54 signaling and suppressed by BMP-4 action in GT1-7 neuron cells.
Collapse
Affiliation(s)
- Tomohiro Terasaka
- Department of Medicine and Clinical Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; Department of General Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Hu J, Du G, Zhang W, Huang H, Chen D, Wu D, Wang X. Short-term neonatal/prepubertal exposure of dibutyl phthalate (DBP) advanced pubertal timing and affected hypothalamic kisspeptin/GPR54 expression differently in female rats. Toxicology 2013; 314:65-75. [DOI: 10.1016/j.tox.2013.09.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2013] [Revised: 09/13/2013] [Accepted: 09/13/2013] [Indexed: 11/29/2022]
|
49
|
Zhang J, Yang L, Lin N, Pan X, Zhu Y, Chen X. Aging-related changes in RP3V kisspeptin neurons predate the reduced activation of GnRH neurons during the early reproductive decline in female mice. Neurobiol Aging 2013; 35:655-68. [PMID: 24112790 DOI: 10.1016/j.neurobiolaging.2013.08.038] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2013] [Revised: 07/23/2013] [Accepted: 08/30/2013] [Indexed: 11/27/2022]
Abstract
Kisspeptin neurons in the rostral periventricular area of the third ventricle (RP3V) play a key role in relaying the positive feedback effects of estradiol that activate gonadotropin-releasing hormone (GnRH) neurons and drive a surge in the GnRH/luteinizing hormone (LH) level. However, the precise role of kisspeptin neurons during female reproductive senescence remains unclear. Focusing on middle-aged intact female mice with irregular estrous cycles, we found a parallel decline in c-Fos-positive kisspeptin neurons and c-Fos-positive GnRH neurons at the time of the GnRH/LH surge. Furthermore, in kisspeptin neurons, the expression of estrogen receptor α (ERα), but not progesterone receptor (PR), decreased with age. Interestingly, some kisspeptin neurons in the RP3V, but none of the GnRH neurons in the rostral preoptic area (rPOA), had a characteristic cellular senescence in middle-aged mice and old mice. These data suggest that, among the groups of neurons involved in reproductive control, the kisspeptin neurons in the RP3V are likely among the earliest to undergo aging processes and thus participate in initiating the early reproductive decline.
Collapse
Affiliation(s)
- Jing Zhang
- Key Laboratory of Brain Aging and Neurodegenerative Disease, Fujian Institute of Geriatrics, The Affiliated Union Hospital of Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China.
| | | | | | | | | | | |
Collapse
|
50
|
Petersen SL, Intlekofer KA, Moura-Conlon PJ, Brewer DN, Del Pino Sans J, Lopez JA. Novel progesterone receptors: neural localization and possible functions. Front Neurosci 2013; 7:164. [PMID: 24065878 PMCID: PMC3776953 DOI: 10.3389/fnins.2013.00164] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 08/24/2013] [Indexed: 12/30/2022] Open
Abstract
Progesterone (P4) regulates a wide range of neural functions and likely acts through multiple receptors. Over the past 30 years, most studies investigating neural effects of P4 focused on genomic and non-genomic actions of the classical progestin receptor (PGR). More recently the focus has widened to include two groups of non-classical P4 signaling molecules. Members of the Class II progestin and adipoQ receptor (PAQR) family are called membrane progestin receptors (mPRs) and include: mPRα (PAQR7), mPRβ (PAQR8), mPRγ (PAQR5), mPRδ (PAQR6), and mPRε (PAQR9). Members of the b5-like heme/steroid-binding protein family include progesterone receptor membrane component 1 (PGRMC1), PGRMC2, neudesin, and neuferricin. Results of our recent mapping studies show that members of the PGRMC1/S2R family, but not mPRs, are quite abundant in forebrain structures important for neuroendocrine regulation and other non-genomic effects of P4. Herein we describe the structures, neuroanatomical localization, and signaling mechanisms of these molecules. We also discuss possible roles for Pgrmc1/S2R in gonadotropin release, feminine sexual behaviors, fluid balance and neuroprotection, as well as catamenial epilepsy.
Collapse
Affiliation(s)
- Sandra L Petersen
- Molecular and Cellular Neuroendocrinology, Department of Veterinary and Animal Sciences, University of Massachusetts Amherst Amherst, MA, USA
| | | | | | | | | | | |
Collapse
|