1
|
Żabińska M, Wiśniewska K, Węgrzyn G, Pierzynowska K. Exploring the physiological role of the G protein-coupled estrogen receptor (GPER) and its associations with human diseases. Psychoneuroendocrinology 2024; 166:107070. [PMID: 38733757 DOI: 10.1016/j.psyneuen.2024.107070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/15/2024] [Accepted: 05/05/2024] [Indexed: 05/13/2024]
Abstract
Estrogen is a group of hormones that collaborate with the nervous system to impact the overall well-being of all genders. It influences many processes, including those occurring in the central nervous system, affecting learning and memory, and playing roles in neurodegenerative diseases and mental disorders. The hormone's action is mediated by specific receptors. Significant roles of classical estrogen receptors, ERα and ERβ, in various diseases were known since many years, but after identifying a structurally and locationally distinct receptor, the G protein-coupled estrogen receptor (GPER), its role in human physiology and pathophysiology was investigated. This review compiles GPER-related information, highlighting its impact on homeostasis and diseases, while putting special attention on functions and dysfunctions of this receptor in neurobiology and biobehavioral processes. Understanding the receptor modulation possibilities is essential for therapy, as disruptions in receptors can lead to diseases or disorders, irrespective of correct estrogen levels. We conclude that studies on the GPER receptor have the potential to develop therapies that regulate estrogen and positively impact human health.
Collapse
Affiliation(s)
- Magdalena Żabińska
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, Gdansk 80-308, Poland
| | - Karolina Wiśniewska
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, Gdansk 80-308, Poland
| | - Grzegorz Węgrzyn
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, Gdansk 80-308, Poland
| | - Karolina Pierzynowska
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, Gdansk 80-308, Poland.
| |
Collapse
|
2
|
Zapater C, Moreira C, Knigge T, Monsinjon T, Gómez A, Pinto PIS. Evolutionary history and functional characterization of duplicated G protein-coupled estrogen receptors in European sea bass. J Steroid Biochem Mol Biol 2024; 236:106423. [PMID: 37939740 DOI: 10.1016/j.jsbmb.2023.106423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 10/29/2023] [Accepted: 11/05/2023] [Indexed: 11/10/2023]
Abstract
Across vertebrates, the numerous estrogenic functions are mainly mediated by nuclear and membrane receptors, including the G protein-coupled estrogen receptor (GPER) that has been mostly associated with rapid non-genomic responses. Although Gper-mediated signalling has been characterized in only few fish species, Gpers in fish appear to present more mechanistic functionalities as those of mammals due to additional gene duplicates. In this study, we ran a thorough investigation of the fish Gper evolutionary history in light of available genomes, we carried out the functional characterization of the two gper gene duplicates of European sea bass (Dicentrarchus labrax) using luciferase reporter gene transactivation assays, validated it with natural and synthetic estrogen agonists/antagonists and applied it to other chemicals of aquaculture and ecotoxicological interest. Phylogenetic and synteny analyses of fish gper1 and gper1-like genes suggest their duplication may have not resulted from the teleost-specific whole genome duplication. We confirmed that both sbsGper isoforms activate the cAMP signalling pathway and respond differentially to distinct estrogenic compounds. Therefore, as observed for nuclear estrogen receptors, both sbsGpers duplicates retain estrogenic activity although they differ in their specificity and potency (Gper1 being more potent and more specific than Gper1-like), suggesting a more conserved role for Gper1 than for Gper1-like. In addition, Gpers were able to respond to estrogenic environmental pollutants known to interfere with estrogen signalling, such as the phytoestrogen genistein and the anti-depressant fluoxetine, a point that can be taken into account in aquatic environment pollution screenings and chemical risk assessment, complementing previous assays for sea bass nuclear estrogen receptors.
Collapse
Affiliation(s)
- Cinta Zapater
- Instituto de Acuicultura Torre de la Sal, CSIC, 12595 Torre de la Sal, Castellón, Spain.
| | - Catarina Moreira
- UMR-I 02 Environmental Stress and Aquatic Biomonitoring (SEBIO), University of Le Havre Normandy, F-76600 Le Havre, France.
| | - Thomas Knigge
- UMR-I 02 Environmental Stress and Aquatic Biomonitoring (SEBIO), University of Le Havre Normandy, F-76600 Le Havre, France.
| | - Tiphaine Monsinjon
- UMR-I 02 Environmental Stress and Aquatic Biomonitoring (SEBIO), University of Le Havre Normandy, F-76600 Le Havre, France.
| | - Ana Gómez
- Instituto de Acuicultura Torre de la Sal, CSIC, 12595 Torre de la Sal, Castellón, Spain.
| | - Patrícia I S Pinto
- Centro de Ciências do Mar (CCMAR), Universidade do Algarve, 8005-139 Faro, Portugal.
| |
Collapse
|
3
|
Wen Y, Zhan J, Li C, Li P, Wang C, Wu J, Xu Y, Zhang Y, Zhou Y, Li E, Nie H, Wu X. G-protein couple receptor (GPER1) plays an important role during ovarian folliculogenesis and early development of the Chinese Alligator. Anim Reprod Sci 2023; 255:107295. [PMID: 37422950 DOI: 10.1016/j.anireprosci.2023.107295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/30/2023] [Accepted: 07/02/2023] [Indexed: 07/11/2023]
Abstract
The critical role of the G protein-coupled receptor 1 (GPER1), a member of the seven-transmembrane G protein-coupled receptor family, in the functional regulation of oocytes accumulated abundant theories in the early research on model animals. However, the full-length cDNA encoding GPER1 and its role in the folliculogenesis has not been illustrated in crocodilians. 0.5, 3, and 12 months old Alligator sinensis cDNA samples were used to clone the full-length cDNA encoding GPER1. Immunolocalization and quantitative analysis were performed using Immunofluorescence technique, RT-PCR and Western blot. Simultaneously, studies on GPER1's promoter deletion and cis-acting transcriptional regulation mechanism were conducted. Immunolocalization staining for the germline marker DDX4 and GPER1 demonstrated that DDX4-positive oocytes were clustered tightly together within the nests, whereas scarcely any detectable GPER1 was present in the oocytes nest in Stage I. After that, occasionally GPER1-positive immunosignal was observed in oocytes and somatic cells additional with the primordial follicles, and it was mainly located at the granulosa cells or thecal cells within the early PFs in the Stage III. The single mutation of the putative SP1 motif, double mutating of Ets/SP1 and SP1/CRE binding sites all depressed promoter activities. This result will help to investigate the role of GPER1 in the early folliculogenesis of A. sinensis.
Collapse
Affiliation(s)
- Yue Wen
- College of Life Sciences, Anhui Normal University, Wuhu, Anhui Province, 241000, People's Republic of China; Key Laboratory for Conservation and Use of Important Biological Resources of Anhui Province, People's Republic of China
| | - Jixiang Zhan
- College of Life Sciences, Anhui Normal University, Wuhu, Anhui Province, 241000, People's Republic of China; Key Laboratory for Conservation and Use of Important Biological Resources of Anhui Province, People's Republic of China
| | - Changcheng Li
- College of Life Sciences, Anhui Normal University, Wuhu, Anhui Province, 241000, People's Republic of China; Key Laboratory for Conservation and Use of Important Biological Resources of Anhui Province, People's Republic of China
| | - Pengfei Li
- College of Life Sciences, Anhui Normal University, Wuhu, Anhui Province, 241000, People's Republic of China; Key Laboratory for Conservation and Use of Important Biological Resources of Anhui Province, People's Republic of China
| | - Chong Wang
- College of Life Sciences, Anhui Normal University, Wuhu, Anhui Province, 241000, People's Republic of China; Key Laboratory for Conservation and Use of Important Biological Resources of Anhui Province, People's Republic of China
| | - Jie Wu
- College of Life Sciences, Anhui Normal University, Wuhu, Anhui Province, 241000, People's Republic of China; Key Laboratory for Conservation and Use of Important Biological Resources of Anhui Province, People's Republic of China
| | - Yunlu Xu
- College of Life Sciences, Anhui Normal University, Wuhu, Anhui Province, 241000, People's Republic of China; Key Laboratory for Conservation and Use of Important Biological Resources of Anhui Province, People's Republic of China
| | - Yuqian Zhang
- College of Life Sciences, Anhui Normal University, Wuhu, Anhui Province, 241000, People's Republic of China; Key Laboratory for Conservation and Use of Important Biological Resources of Anhui Province, People's Republic of China
| | - Yongkang Zhou
- Alligator Research Center of Anhui Province, Xuanzhou 242000, People's Republic of China
| | - En Li
- College of Life Sciences, Anhui Normal University, Wuhu, Anhui Province, 241000, People's Republic of China; Key Laboratory for Conservation and Use of Important Biological Resources of Anhui Province, People's Republic of China
| | - Haitao Nie
- College of Life Sciences, Anhui Normal University, Wuhu, Anhui Province, 241000, People's Republic of China; Key Laboratory for Conservation and Use of Important Biological Resources of Anhui Province, People's Republic of China.
| | - Xiaobing Wu
- College of Life Sciences, Anhui Normal University, Wuhu, Anhui Province, 241000, People's Republic of China; Key Laboratory for Conservation and Use of Important Biological Resources of Anhui Province, People's Republic of China.
| |
Collapse
|
4
|
Takahashi T, Ogiwara K. cAMP signaling in ovarian physiology in teleosts: A review. Cell Signal 2023; 101:110499. [PMID: 36273754 DOI: 10.1016/j.cellsig.2022.110499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/11/2022] [Accepted: 10/15/2022] [Indexed: 11/30/2022]
Abstract
Ovarian function in teleosts, like in other vertebrates, is regulated by two distinct gonadotropins, follicle-stimulating hormone (FSH) and luteinizing hormone (LH). Gonadotropin effects are mediated by membrane-bound G protein-coupled receptors localized on the surface of follicle cells. Gonadotropin receptor activation results in increased intracellular cAMP, the most important second cellular signaling molecule. FSH stimulation induces the production of 17β-estradiol in the cells of growing follicles to promote vitellogenesis in oocytes. In contrast, in response to LH, fully grown post-vitellogenic follicles gain the ability to synthesize maturation-inducing steroids, which induce meiotic resumption and ovulation. All these events were induced downstream of cAMP. In this review, we summarize studies addressing the role of the cAMP pathway in gonadotropin-induced processes in teleost ovarian follicles. Furthermore, we discuss future problems concerning cAMP signaling in relation to teleost ovarian function and the differences and similarities in the gonadotropin-induced cAMP signaling pathways between mammals and teleosts.
Collapse
Affiliation(s)
- Takayuki Takahashi
- Laboratory of Reproductive and Developmental Biology, Faculty of Science, Hokkaido University, Sapporo 060-0810, Japan
| | - Katsueki Ogiwara
- Laboratory of Reproductive and Developmental Biology, Faculty of Science, Hokkaido University, Sapporo 060-0810, Japan.
| |
Collapse
|
5
|
Xu XL, Huang ZY, Yu K, Li J, Fu XW, Deng SL. Estrogen Biosynthesis and Signal Transduction in Ovarian Disease. Front Endocrinol (Lausanne) 2022; 13:827032. [PMID: 35299973 PMCID: PMC8921451 DOI: 10.3389/fendo.2022.827032] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/03/2022] [Indexed: 12/01/2022] Open
Abstract
Estrogen mainly binds to estrogen receptors (ERs) to regulate menstrual cycles and reproduction. The expression of ERalpha (ERα), ERbeta (ERβ), and G-protein-coupled estrogen receptor (GPER) mRNA could be detected in ovary, suggesting that they play an important role in estrogen signal transduction in ovary. And many studies have revealed that abnormal expression of estrogen and its receptors is closely related to ovarian disease or malignant tumors. With the continuous development and research of animal models, tissue-specific roles of both ERα and ERβ have been demonstrated in animals, which enable people to have a deeper understanding of the potential role of ER in regulating female reproductive diseases. Nevertheless, our current understanding of ERs expression and function in ovarian disease is, however, incomplete. To elucidate the biological mechanism behind ERs in the ovary, this review will focus on the role of ERα and ERβ in polycystic ovary syndrome (PCOS), ovarian cancer and premature ovarian failure (POF) and discuss the major challenges of existing therapies to provide a reference for the treatment of estrogen target tissue ovarian diseases.
Collapse
Affiliation(s)
- Xue-Ling Xu
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zheng-Yuan Huang
- Department of Metabolism, Digestion and Reproduction, Chelsea and Westminster Hospital, Imperial College London, London, United Kingdom
| | - Kun Yu
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jun Li
- Department of Reproductive Medicine, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiang-Wei Fu
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shou-Long Deng
- National Health Commission of China (NHC) Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| |
Collapse
|
6
|
Abstract
Steroid hormones bind receptors in the cell nucleus and in the cell membrane. The most widely studied class of steroid hormone receptors are the nuclear receptors, named for their function as ligand-dependent transcription factors in the cell nucleus. Nuclear receptors, such as estrogen receptor alpha, can also be anchored to the plasma membrane, where they respond to steroids by activating signaling pathways independent of their function as transcription factors. Steroids can also bind integral membrane proteins, such as the G protein-coupled estrogen receptor. Membrane estrogen and progestin receptors have been cloned and characterized in vitro and influence the development and function of many organ systems. Membrane androgen receptors were cloned and characterized in vitro, but their function as androgen receptors in vivo is unresolved. We review the identity and function of membrane proteins that bind estrogens, progestins, and androgens. We discuss evidence that membrane glucocorticoid and mineralocorticoid receptors exist, and whether glucocorticoid and mineralocorticoid nuclear receptors act at the cell membrane. In many cases, integral membrane steroid receptors act independently of nuclear steroid receptors, even though they may share a ligand.
Collapse
Affiliation(s)
- Lindsey S Treviño
- Department of Population Sciences, Division of Health Equities, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Daniel A Gorelick
- Center for Precision Environmental Health, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Correspondence: Daniel A Gorelick, PhD, One Baylor Plaza, Alkek Building N1317.07, Houston, TX, 77030-3411, USA.
| |
Collapse
|
7
|
Meng F, Zhang L, Zhang W. Two forms of G protein-coupled estrogen receptor 1 in the ricefield eel: Expression and functional characterization in relation to ovarian follicle development. Gen Comp Endocrinol 2021; 304:113720. [PMID: 33508329 DOI: 10.1016/j.ygcen.2021.113720] [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: 09/06/2020] [Revised: 01/01/2021] [Accepted: 01/15/2021] [Indexed: 01/02/2023]
Abstract
G protein-coupled estrogen receptor 1 (Gper1) mediates many rapid, non-genomic estrogenic effects in vertebrates, and plays an important reproductive role in the maintenance of oocyte meiotic arrest in teleost fishes. In the present study, two genes for Gper1, namely gper1a and gper1b, were identified in the genome of a teleost fish, the ricefield eel (Monopterus albus) through Blast and syntenic analysis. Although genes neighboring gper1b are of high synteny, ricefield eel Gper1b shares very low (around 15) percent identities with Gper1 homologues of other vertebrates. In transiently transfected HEK293T cells, both ricefield eel Gper1a and Gper1b responded to estradiol (E2) and estradiol-BSA (E2-BSA) challenges by activating pCRE but not pSRE luciferase reporters, which were abolished by G-15 and NF-449. The production of cAMP was also increased in HEK293T cells transfected with Gper1a or Gper1b expression construct after E2-BSA challenge, which was also abolished by G-15. Surprisingly, both Gper1a and Gper1b showed ligand-independent effects on pCRE luciferase reporters at higher transfected doses (10 ng). RT-PCR analysis showed that the transcript of gper1a is broadly expressed in tissues of both female and male fish while the expression of gper1b in tissues demonstrates obvious sexual dimorphism, with transcripts detected in all tissues examined in female whereas they were barely detectable in some peripheral tissues of male including the testis. In the ovary, the expression of both gper1a and gper1b was detected in the oocyte but not the follicular layer, with the mRNA levels increased during vitellogenesis, peaked at the late vitellogenic stage, and decreased precipitously at the full-grown and germinal vesicle breakdown (GVBD) stages. Moreover, E2 and E2-BSA induced cAMP production in the in vitro incubated follicles at mid-vitellogenic stage but not the GVBD stage, and the induction could be completely abolished by G-15, a Gper1 inhibitor. Taken together, these results suggest that both Gper1a and Gper1b may play important roles in the development and maturation of ovarian follicles in ricefield eels, possibly through inhibition of oocyte meiotic resumption.
Collapse
Affiliation(s)
- Feiyan Meng
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou, PR China
| | - Lihong Zhang
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou, PR China; Biology Department, School of Life Sciences, Sun Yat-Sen University, Guangzhou, PR China.
| | - Weimin Zhang
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou, PR China; Biology Department, School of Life Sciences, Sun Yat-Sen University, Guangzhou, PR China.
| |
Collapse
|
8
|
Cao M, Wang Y, Yang F, Li J, Qin X. Melatonin rescues the reproductive toxicity of low-dose glyphosate-based herbicide during mouse oocyte maturation via the GPER signaling pathway. J Pineal Res 2021; 70:e12718. [PMID: 33503294 DOI: 10.1111/jpi.12718] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 12/16/2020] [Accepted: 01/22/2021] [Indexed: 01/01/2023]
Abstract
Glyphosate-based herbicides (GBHs) are a group of widely used broad-spectrum agricultural pesticides. Due to the recalcitrance of GBH, it has been found in food and environment as a contaminant, posing a threat to public health. The health risks associated with GBH have been indicated by reporting acute toxicity data (an acute exposure of GBH at a 0.5% dose), which primarily discuss toxicity in relation to accidental high-rate exposure. Currently, there is little information regarding the toxicity of GBH at environmentally relevant levels. In this study, we used mature mouse oocytes to study the toxic effects of low-dose GBH exposure in vitro (0.00001%-0.00025%) and in vivo (0.0005%, orally administered through daily drinking water) during meiotic maturation. GBH exposure led to meiotic maturation failure with spindle defects and chromosome misalignment. In addition, GBH treatment severely reduced sperm-binding ability and disrupted early embryo cleavage. Moreover, GBH exposure significantly increased the reactive oxygen species (ROS) levels and apoptotic rates. Evidence indicates that such effects in GBH-exposed oocytes are likely due to overexpression of the G-protein estrogen receptor (GPER/GPR30). Remarkably, we found that melatonin administration elicited significant protection against GBH-induced oocyte deterioration via preserving the expression of GPR30, along with activation of its downstream signaling event (pERK/ERK). Taken together, these results revealed that low-dose glyphosate has a certain adverse effect on oocyte maturation and early embryo cleavage, and highlight the protective roles of melatonin.
Collapse
Affiliation(s)
- Mingjun Cao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yufeng Wang
- Ministry of Education and Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Fan Yang
- Bureau of Agriculture and Rural Affairs of Hanting District, Weifang, China
| | - Jizhou Li
- Joint Graduate Program of Peking-Tsinghua-NIBS, School of Life Sciences, Tsinghua University, Beijing, China
| | - Xunsi Qin
- Joint Graduate Program of Peking-Tsinghua-NIBS, School of Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
9
|
Zhang D, Xia T, Li H, Li Z, Sun G, Li G, Tian Y, Liu X, Xu D, Kang X. Estrogen enhances the expression of a growth-associated long noncoding RNA in chicken liver via ERα. Br Poult Sci 2021; 62:336-345. [PMID: 33390024 DOI: 10.1080/00071668.2020.1868405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
1. The long noncoding RNA lncGLM is significantly differentially expressed in the livers of peak-laying hens compared with that in the livers of pre-laying hens, but its potential biological role and expression regulation are unclear.2. To explore the potential biological function of lncGLM, single nucleotide polymorphism (SNP) detection and association analysis were carried out in the Gushi×Anka F2 resource population.3. The tissues and spatiotemporal expression characteristics of lncGLM were analysed by real-time quantitative PCR. The effects of 17β-oestradiol on the expression of lncGLM expression were analysed through in vitro and in vivo experiments.4. The results showed that a g.19069338 T > C SNP was present in lncGLM. Association analysis revealed that lncGLM was significantly associated with body slanting length at 12 weeks, body weight at 12 weeks, shank length at four weeks, chest depth at eight weeks, pelvic width at 12 weeks, eviscerated weight, head weight, pancreas weight, pectoralis weight, leg muscle weight, muscular stomach weight rate, pancreas weight rate, carcase weight, aspartate aminotransferase, creatinine and pectoral muscle water loss rate.5. The expression of lncGLM in the liver was higher than that in other sampled tissues. In addition, the expression of lncGLM in the liver was significantly higher in the peak-laying period than at the pre-laying period. Both in vitro and in vivo experiments showed that lncGLM expression was regulated by 17β-oestradiol via oestrogen receptor alpha (ER-α). These results demonstrated that the chicken lncGLM gene is highly expressed in liver tissue and regulated by oestrogen through ER-α.
Collapse
Affiliation(s)
- D Zhang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - T Xia
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - H Li
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China.,Henan Innovative Engineering Research Center of Poultry Germplasm Resource, Henan Agricultural University, Zhengzhou, China.,International Joint Research Laboratory for Poultry Breeding of Henan, Henan Agricultural University, Zhengzhou, China
| | - Z Li
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China.,Henan Innovative Engineering Research Center of Poultry Germplasm Resource, Henan Agricultural University, Zhengzhou, China.,International Joint Research Laboratory for Poultry Breeding of Henan, Henan Agricultural University, Zhengzhou, China
| | - G Sun
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China.,Henan Innovative Engineering Research Center of Poultry Germplasm Resource, Henan Agricultural University, Zhengzhou, China.,International Joint Research Laboratory for Poultry Breeding of Henan, Henan Agricultural University, Zhengzhou, China
| | - G Li
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China.,Henan Innovative Engineering Research Center of Poultry Germplasm Resource, Henan Agricultural University, Zhengzhou, China.,International Joint Research Laboratory for Poultry Breeding of Henan, Henan Agricultural University, Zhengzhou, China
| | - Y Tian
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China.,Henan Innovative Engineering Research Center of Poultry Germplasm Resource, Henan Agricultural University, Zhengzhou, China.,International Joint Research Laboratory for Poultry Breeding of Henan, Henan Agricultural University, Zhengzhou, China
| | - X Liu
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China.,Henan Innovative Engineering Research Center of Poultry Germplasm Resource, Henan Agricultural University, Zhengzhou, China.,International Joint Research Laboratory for Poultry Breeding of Henan, Henan Agricultural University, Zhengzhou, China
| | - D Xu
- Henan Liujiang Ecological Animal Husbandry Co., Ltd, Hebi, China
| | - X Kang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China.,Henan Innovative Engineering Research Center of Poultry Germplasm Resource, Henan Agricultural University, Zhengzhou, China.,International Joint Research Laboratory for Poultry Breeding of Henan, Henan Agricultural University, Zhengzhou, China
| |
Collapse
|
10
|
Miyaoku K, Ogino Y, Lange A, Ono A, Kobayashi T, Ihara M, Tanaka H, Toyota K, Akashi H, Yamagishi G, Sato T, Tyler CR, Iguchi T, Miyagawa S. Characterization of G protein-coupled estrogen receptors in Japanese medaka, Oryzias latipes. J Appl Toxicol 2020; 41:1390-1399. [PMID: 33336402 DOI: 10.1002/jat.4130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 12/03/2020] [Accepted: 12/03/2020] [Indexed: 11/06/2022]
Abstract
The G protein-coupled estrogen receptor 1 (Gper1) is a membrane-bound estrogen receptor that mediates non-genomic action of estrogens. A Gper1-mediating pathway has been implicated in reproductive activities in fish, including oocyte growth, but Gper1 has been characterized in only a very limited number of fish species. In this study, we cloned and characterized two genes encoding medaka (Oryzias latipes) Gper1s, namely, Gper1a and Gper1b, and phylogenic and synteny analyses suggest that these genes originate through a teleost-specific whole genome duplication event. We found that Gper1a induced phosphorylation of mitogen-activated protein kinase (MAPK) in 293T cells transfected with medaka Gper1s on exposure to the natural estrogen, 17β-estradiol (E2) and a synthetic Gper1 agonist (G-1), and treatment with both E2 and G-1 also decreased the rate of spontaneous maturation in medaka oocytes. These findings show that the processes for oocyte growth and maturation are sensitive to estrogens and are possibly mediated through Gper1a in medaka. We also show that 17α-ethinylestradiol (EE2), one of the most potent estrogenic endocrine-disrupting chemicals, and bisphenol A (BPA, a weak environmental estrogen) augmented phosphorylation of MAPK through medaka Gper1s in 293T cells. Interestingly, however, treatment with EE2 or BPA did not attenuate maturation of medaka oocytes. Our findings support that Gper1-mediated effects on oocytes are conserved among fish species, but effects of estrogenic endocrine-disrupting chemicals on oocytes acting through Gper1 may be divergent among fish species.
Collapse
Affiliation(s)
- Kaori Miyaoku
- Faculty of Industrial Science and Technology, Tokyo University of Science, Tokyo, Japan
| | - Yukiko Ogino
- Center for Promotion of International Education and Research, Faculty of Agriculture, Kyushu University, Fukuoka, Japan
| | - Anke Lange
- Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, UK
| | - Ayaka Ono
- Faculty of Industrial Science and Technology, Tokyo University of Science, Tokyo, Japan
| | - Tohru Kobayashi
- Graduate School of Nutritional and Environmental Sciences, University of Shizuoka, Shizuoka, Japan
| | - Masaru Ihara
- Research Centre for Environmental Quality Management, Kyoto University, Ohtsu, Japan
| | - Hiroaki Tanaka
- Research Centre for Environmental Quality Management, Kyoto University, Ohtsu, Japan
| | - Kenji Toyota
- Faculty of Industrial Science and Technology, Tokyo University of Science, Tokyo, Japan.,Sado Marine Biological Station, Faculty of Science, Niigata University, Sado, Japan
| | - Hiroshi Akashi
- Faculty of Industrial Science and Technology, Tokyo University of Science, Tokyo, Japan
| | - Genki Yamagishi
- Faculty of Industrial Science and Technology, Tokyo University of Science, Tokyo, Japan
| | - Tomomi Sato
- Graduate School of Nanobioscience, Yokohama City University, Yokohama, Japan
| | - Charles R Tyler
- Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, UK
| | - Taisen Iguchi
- Graduate School of Nanobioscience, Yokohama City University, Yokohama, Japan
| | - Shinichi Miyagawa
- Faculty of Industrial Science and Technology, Tokyo University of Science, Tokyo, Japan.,Division of Biological Environment Innovation, Research Institute for Science and Technology, Tokyo University of Science, Tokyo, Japan
| |
Collapse
|
11
|
Jeng SR, Thomas P, Pang Y, Dufour S, Lin CJ, Yueh WS, Chang CF. Elevated estradiol-17β levels inhibit final oocyte maturation via G protein-coupled estrogen receptor (Gper) in yellowfin porgy, Acanthopagrus latus. Gen Comp Endocrinol 2020; 299:113587. [PMID: 32827512 DOI: 10.1016/j.ygcen.2020.113587] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 08/07/2020] [Accepted: 08/13/2020] [Indexed: 02/06/2023]
Abstract
Yellowfin porgy a protandrous teleost, exhibits asynchronous oocyte development and multiple spawning. Seasonal profiles of plasma estradiol-17β (E2) levels showed a peak in three-year-old females during the spawning season, when batches of fully-grown oocytes undergo final oocyte maturation (FOM). Because E2 has been shown to inhibit FOM via the G protein-coupled estrogen receptor (Gper) in several teleost species, we investigated the role of this "paradoxical" increase in E2 during FOM in yellowfin porgy. In vivo treatment with a GnRH-agonist stimulated germinal vesicle breakdown (GVBD) and increased E2 plasma levels, and ovarian cyp19a1a transcripts, confirming the increase in E2 production at the time of FOM. Ovarian transcripts of gper peaked at the time of FOM, indicating an increase in ovarian responsiveness to Gper-mediated E2 effects. In vitro, E2 and the Gper agonist, G-1, inhibited the stimulatory effect of maturation-inducing steroids (MIS) on GVBD, while an aromatase inhibitor enhanced the MIS effect, in agreement with a physiological inhibitory role of E2 on FOM via Gper. Immunohistological studies showed that the Gper protein was specifically located on the oocyte plasma membrane. Ovarian membranes displayed high-affinity and limited-capacity specific [3H]-E2 receptor binding which was displaced by G-1, characteristic of Gper. Expression of gper increased at the time of FOM in mid-vitellogenic oocytes, but not in larger oocytes undergoing GVBD. These results suggest increases in both E2 production and E2 responsiveness via Gper upregulation in mid-vitellogenic oocytes, may maintain meiotic arrest in this oocyte stage class during the period when full-grown oocytes are undergoing FOM. This study indicates a critical involvement of E2 in the control of asynchronous oocyte maturation and the multiple spawning pattern in Sparidae.
Collapse
Affiliation(s)
- Shan-Ru Jeng
- Department of Aquaculture, National Kaohsiung University of Science and Technology, Kaohsiung 81157, Taiwan
| | - Peter Thomas
- Marine Science Institute, University of Texas at Austin, Port Aransas, TX 78373, USA
| | - Yefei Pang
- Marine Science Institute, University of Texas at Austin, Port Aransas, TX 78373, USA
| | - Sylvie Dufour
- Laboratory Biology of Aquatic Organisms and Ecosystems (BOREA), Muséum National d'Histoire Naturelle, CNRS, IRD, Sorbonne Université, Université de Caen Normandie, Université des Antilles, 75231 Paris Cedex 05, France
| | - Chien-Ju Lin
- Department of Aquaculture, National Taiwan Ocean University, Keelung 20224, Taiwan; Center of Excellence for the Oceans, National Taiwan Ocean University, Keelung 20224, Taiwan
| | - Wen-Shiun Yueh
- Department of Aquaculture, National Kaohsiung University of Science and Technology, Kaohsiung 81157, Taiwan.
| | - Ching-Fong Chang
- Department of Aquaculture, National Taiwan Ocean University, Keelung 20224, Taiwan; Center of Excellence for the Oceans, National Taiwan Ocean University, Keelung 20224, Taiwan.
| |
Collapse
|
12
|
Converse A, Thomas P. Androgens regulate follicle stage-dependent pro- and anti-apoptosis in teleost ovaries through ZIP9 activation of different G proteins†. Biol Reprod 2020; 101:377-391. [PMID: 31074766 DOI: 10.1093/biolre/ioz086] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 04/24/2019] [Accepted: 05/09/2019] [Indexed: 12/18/2022] Open
Abstract
Androgens mediate a number of processes in mammalian and teleost ovaries in a follicle-stage dependent manner, including follicle growth, survival, and apoptosis. We recently reported that the membrane androgen receptor ZIP9 mediates apoptosis in Atlantic croaker granulosa/theca (G/T) cells from mature ovarian follicles, but the effects of androgens on early stage G/T cells in this model remains unknown. Here we show that testosterone mediates pro- and anti-apoptotic responses in a follicle stage-dependent manner in croaker ovarian follicle cells. Testosterone treatment decreased the incidence of apoptosis in G/T cells from early stage follicles (diameter <300 μm) but increased apoptosis in G/T cells from late stage follicles (diameter >400 μm). Small interfering RNA targeting ZIP9, but not the nuclear androgen receptor, blocked the anti-apoptotic response, indicating ZIP9 mediates anti-apoptotic in addition to pro-apoptotic responses. Testosterone treatment of early stage G/T cells resulted in opposite signaling outcomes from those previously characterized for the ZIP9-mediated apoptotic response including decreased cAMP and intracellular free zinc levels, and downregulation of pro-apoptotic member mRNA expression. While ZIP9-mediated apoptosis involves activation of a stimulatory G protein (Gs), activators of Gs signaling antagonized the anti-apoptotic response. Proximity ligation and G protein activation assays indicated that in G/T cells from early stage follicles ZIP9 is in close proximity and activates an inhibitory G protein, while in G/T cells from late stage follicles ZIP9 is in close proximity and activates Gs. This study demonstrates that ZIP9 mediates opposite survival responses of croaker G/T cells by activating different G proteins in a follicle stage-dependent manner.
Collapse
Affiliation(s)
- Aubrey Converse
- Marine Science Institute, The University of Texas at Austin, Port Aransas, Texas, USA
| | - Peter Thomas
- Marine Science Institute, The University of Texas at Austin, Port Aransas, Texas, USA
| |
Collapse
|
13
|
Li H, Li Y, Yang L, Zhang D, Liu Z, Wang Y, Han R, Li G, Li Z, Tian Y, Kang X, Liu X. Identification of a Novel Lipid Metabolism-Associated Hepatic Gene Family Induced by Estrogen via ERα in Chicken ( Gallus gallus). Front Genet 2020; 11:271. [PMID: 32296460 PMCID: PMC7136477 DOI: 10.3389/fgene.2020.00271] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 03/06/2020] [Indexed: 12/17/2022] Open
Abstract
Liver is the main organ of lipid metabolism in chicken, especially for laying hens. To explore the molecular mechanism of lipid metabolism in chicken, five novel genes discovered in chicken liver tissue were systematically studied. Bioinformatic analysis was used to analyze the gene characteristics. The expression patterns and regulatory molecular mechanism of the five genes were examined. Our results showed that all five novel genes contain a common NADP-binding site that belongs to the NADB-Rossmann superfamily, and the genes were designated NADB-LER1-5. Phylogenetic tree of the NADB-LERs gene family in different species suggested these five genes originated from the same ancestor. Tissue distributions showed that NADB-LER1-4 genes were highly expressed in lipid metabolism organs, including liver, kidney and duodenum, and that the NADB-LER5 gene was highly expressed in liver and kidney. The spatiotemporal expression indicated that the expression levels of NADB-LER1-5 genes in liver tissue were significantly greater in sexually mature hens than that of immature pullets (P-value ≤ 0.05). The expression levels of NADB-LER1-5 were significantly induced by 17β-estradiol in primary cultured chicken embryo hepatocytes (P-value ≤ 0.05), and 17β-estradiol regulated the expression of NADB-LER1-5 mediated by ERα. Individual assays verified that under induction of 17β-estradiol, the five novel genes were significantly upregulated, with subsequent alteration in serum TG, TC, and VLDLs in 10-week-old pullets. This study proved NADB-LERs family mainly expressed in liver, kidney, and duodenum tissues. 17β-estradiol induces the expression of NADB-LER1-5 genes predominantly mediated via ERα. They likely involved in lipid metabolism in the liver of chicken.
Collapse
Affiliation(s)
- Hong Li
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Yanmin Li
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Liyu Yang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Dingding Zhang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Ziming Liu
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Yanbin Wang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Ruili Han
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Guoxi Li
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Zhuanjian Li
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Yadong Tian
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Xiangtao Kang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Xiaojun Liu
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| |
Collapse
|
14
|
Evans PD. Rapid signalling responses via the G protein-coupled estrogen receptor, GPER, in a hippocampal cell line. Steroids 2019; 152:108487. [PMID: 31499073 DOI: 10.1016/j.steroids.2019.108487] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 08/19/2019] [Accepted: 09/03/2019] [Indexed: 01/14/2023]
Abstract
The rapid non-genomic actions of 17β-estradiol in multiple tissues, including the nervous system, may involve the activation of the G-protein-coupled receptor, GPER. Different signalling pathways have been suggested to be activated by GPER in different cell lines and tissues. Controversially, GPER has also been suggested to be activated by the mineralocorticoid aldosterone, and by the non-steroidal diphenylacrylamide compound, STX, in some preparations. Evidence for the ability of the GPER agonist, G-1, and for aldosterone in the presence of the mineralocorticoid receptor antagonist, eplerenone, to potentiate forskolin-stimulated cyclic AMP levels in the hippocampal clonal cell line, mHippoE-18 is reviewed. The effects of both agents are blocked by the GPER antagonist G36, by PTX, (suggesting the involvement of Gi/o G proteins), by BAPTA-AM, (suggesting they are calcium sensitive), by wortmannin (suggesting an involvement of PI3Kinase) and by soluble amyloid-β peptides. STX also stimulates cyclic AMP levels in mHippoE-18 cells and these effects are blocked by G36 and PTX, as well as by amyloid-β peptides. This suggests that both aldosterone and STX may be capable of activating GPER in mHippoE-18 cells. Possible molecular mechanisms that may underlie these effects are discussed, together with possible forward directions for research on rapid non-genomic signalling by GPER, emphasising the importance of understanding the spatio-temporal aspects of its signalling in various tissues.
Collapse
Affiliation(s)
- Peter D Evans
- The Signalling Laboratory, The Babraham Institute, The Babraham Research Campus, Cambridge CB22 3AT, UK.
| |
Collapse
|
15
|
Evans PD. Aldosterone, STX and amyloid-β 1-42 peptides modulate GPER (GPR30) signalling in an embryonic mouse hippocampal cell line (mHippoE-18). Mol Cell Endocrinol 2019; 496:110537. [PMID: 31404576 DOI: 10.1016/j.mce.2019.110537] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 08/05/2019] [Accepted: 08/08/2019] [Indexed: 01/02/2023]
Abstract
The GPCR, GPER, mediates many of the rapid, non-genomic actions of 17β-estradiol in multiple tissues, including the nervous system. Controversially, it has also been suggested to be activated by aldosterone, and by the non-steroidal diphenylacrylamide compound, STX, in some preparations. Here, the ability of the GPER agonist, G-1, and aldosterone in the presence of the mineralocorticoid receptor antagonist, eplerenone, to potentiate forskolin-stimulated cyclic AMP levels in the hippocampal clonal cell line, mHippoE-18, are compared. Both stimulatory effects are blocked by the GPER antagonist G36, by PTX, (suggesting the involvement of Gi/o G proteins), by BAPTA-AM, (suggesting they are calcium sensitive), by wortmannin (suggesting an involvement of PI3Kinase) and by soluble amyloid-β peptides. STX also stimulates cyclic AMP levels in mHippoE-18 cells and these effects are blocked by G36 and PTX, as well as by amyloid-β peptides. This suggests that both aldosterone and STX may modulate GPER signalling in mHippoE-18 cells.
Collapse
Affiliation(s)
- Peter D Evans
- The Signalling Laboratory, The Babraham Institute, The Babraham Research Campus, Cambridge, CB22 3AT, UK.
| |
Collapse
|
16
|
Zhang H, Wei Q, Gao Z, Ma C, Yang Z, Zhao H, Liu C, Liu J, Zhao X, Ma B. G protein-coupled receptor 30 mediates meiosis resumption and gap junction communications downregulation in goat cumulus-oocyte complexes by 17β-estradiol. J Steroid Biochem Mol Biol 2019; 187:58-67. [PMID: 30414946 DOI: 10.1016/j.jsbmb.2018.11.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/18/2018] [Accepted: 11/03/2018] [Indexed: 11/17/2022]
Abstract
Estrogen plays a critical role in the regulation of gap junctions between oocytes and granulosa cells in mammalian ovaries. G protein-coupled receptor 30 (GPR30) was identified as a membrane estrogen receptor, mediating rapid, nongenomic signaling events that might be responsible for the regulation of oocyte meiosis resumption and gap junction intercellular communications (GJICs). The present study aimed to determine the expression and localization of GPR30 and its role in oocyte meiotic progression and GJICs in goat cumulus-oocyte complexes (COCs). Immunofluorescence experiments revealed that GPR30 was primarily located in the plasma membrane of cumulus cells and oocytes in goats. 17β-estradiol could promote oocyte meiotic progression, which was blocked by G15 (a selective GPR30 antagonist) but not ICI182780 (a nuclear estrogen receptor inhibitor) in the early stage of in vitro culture. The effect of 17β-estradiol on the GJICs was quantified by lucifer yellow (LY) microinjection and calcein-AM fluorescent dye diffusion. 17β-estradiol treatment of goat COCs resulted in rapid downregulation of GJICs. The transfer of calcein from cumulus cells to oocytes could be significantly inhibited by carbenoxolone (a known gap junction blocker), 17β-estradiol or G1 (a GPR30 agonist), and this inhibition could be reversed by G15 but not ICI182780, indicating that GPR30 mediates the effect of 17β-estradiol on the rapid downregulation of GJICs. 17β-estradiol also stimulated the serine 368 phosphorylation of connexin 43 (Cx43) when COCs were in vitro cultured for 4 h, 6 h, and 8 h. More importantly, 17β-estradiol or G1 could separately promote the phosphorylation of extracellular signal-regulated kinase 1/2 (ERK 1/2) and Cx43 significantly when COCs were cultured for 4 h. Furthermore, both ERK1/2 and Cx43 phosphorylation could be inhibited by G15 and the epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor AG1478 or by the ERK1/2 inhibitor PD98059, indicating that EGFR-ERK1/2 signaling was involved in these events. These results supported the hypothesis that GPR30 mediated 17β-estradiol-stimulated meiotic resumption and GJIC reduction in goat COCs. Thus, the present study provides novel insights into elucidating the mechanisms for steroid hormone action in the regulation of oocyte maturation.
Collapse
Affiliation(s)
- Hui Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People's Republic of China
| | - Qiang Wei
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People's Republic of China
| | - Zhen Gao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People's Republic of China
| | - Chiyuan Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People's Republic of China
| | - Zhenshan Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People's Republic of China
| | - Hui Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People's Republic of China
| | - Chen Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People's Republic of China
| | - Jie Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People's Republic of China
| | - Xiaoe Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People's Republic of China
| | - Baohua Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People's Republic of China.
| |
Collapse
|
17
|
Zimmerman MA, Hutson DD, Mauvais-Jarvis F, Lindsey SH. Bazedoxifene-induced vasodilation and inhibition of vasoconstriction is significantly greater than estradiol. Menopause 2019; 26:172-181. [PMID: 30130290 PMCID: PMC6344253 DOI: 10.1097/gme.0000000000001195] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVE A new strategy for menopausal hormone therapy replaces medroxyprogesterone with the selective estrogen receptor modulator bazedoxifene. While the agonist or antagonist activity of bazedoxifene has been examined in other tissues, the current study explored the impact of bazedoxifene on resistance artery reactivity. We hypothesized that bazedoxifene may induce greater vasoprotective effects than estradiol due to enhanced activation of the G-protein-coupled estrogen receptor. METHODS We measured the vasodilation of mesenteric resistance arteries from adult male and female wild-type and G-protein-coupled estrogen receptor knockout mice (n = 58) in response to increasing concentrations of bazedoxifene, medroxyprogesterone, and estradiol, and also the impact of these compounds on the responses to phenylephrine and sodium nitroprusside. RESULTS Bazedoxifene-induced vasorelaxation was greater than estradiol and blunted phenylephrine-induced contraction-an effect not observed with estradiol. Neither estradiol nor bazedoxifene altered relaxation to sodium nitroprusside. The combination of bazedoxifene + estradiol promoted greater vasodilation than medroxyprogesterone + estradiol, and opposed phenylephrine-induced contraction, whereas medroxyprogesterone + estradiol failed to attenuate this response. Both bazedoxifene + estradiol and medroxyprogesterone + estradiol enhanced sodium nitroprusside-induced relaxation in females. Vascular responses were similar in both sexes in wild-type and G-protein-coupled estrogen receptor knockout mice. CONCLUSION Bazedoxifene and bazedoxifene + estradiol relaxed mesenteric arteries and opposed vasoconstriction to a greater degree than estradiol or medroxyprogesterone + estradiol. These effects were independent of sex and G-protein-coupled estrogen receptor expression. We conclude that bazedoxifene may provide vascular benefits over estrogen alone or estrogen plus progestogen combinations in postmenopausal women.
Collapse
Affiliation(s)
- Margaret A Zimmerman
- Department of Pharmacology, Section of Endocrinology, Tulane University School of Medicine, New Orleans, LA
| | - Dillion D Hutson
- Department of Pharmacology, Section of Endocrinology, Tulane University School of Medicine, New Orleans, LA
| | - Franck Mauvais-Jarvis
- Department of Medicine, Section of Endocrinology, Tulane University School of Medicine, New Orleans, LA
| | - Sarah H Lindsey
- Department of Pharmacology, Section of Endocrinology, Tulane University School of Medicine, New Orleans, LA
| |
Collapse
|
18
|
DeCourten BM, Connon RE, Brander SM. Direct and indirect parental exposure to endocrine disruptors and elevated temperature influences gene expression across generations in a euryhaline model fish. PeerJ 2019; 7:e6156. [PMID: 30643694 PMCID: PMC6329337 DOI: 10.7717/peerj.6156] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 11/24/2018] [Indexed: 12/22/2022] Open
Abstract
Aquatic organisms inhabiting polluted waterways face numerous adverse effects, including physiological disruption by endocrine disrupting compounds (EDCs). Little is known about how the temperatures associated with global climate change may influence the response of organisms exposed to EDCs, and the effects that these combined stressors may have on molecular endpoints such as gene expression. We exposed Menidia beryllina (inland silversides) to environmentally relevant concentrations (1 ng/L) of two estrogenic EDCs (bifenthrin and 17α-ethinylestradiol; EE2) at 22 °C and 28 °C. We conducted this experiment over multiple generations to better understand the potential effects to chronically exposed populations in the wild. We exposed adult parental fish (F0) for 14 days prior to spawning of the next generation. F1 larvae were then exposed from fertilization until 21 days post hatch (dph) before being transferred to clean water tanks. F1 larvae were reared to adulthood, then spawned in clean water to test for further effects of parental exposure on offspring (F2 generation). Gene expression was quantified by performing qPCR on F0 and F1 gonads, as well as F1 and F2 larvae. We did not detect any significant differences in the expression of genes measured in the parental or F1 adult gonads. We found that the 28 °C EE2 treatment significantly decreased the expression of nearly all genes measured in the F1 larvae. This pattern was transferred to the F2 generation for expression of the follicle-stimulating hormone receptor (FSHR) gene. Expression of 17β-hydroxysteroid dehydrogenase (17β-HSD) and G protein-coupled receptor 30 (GPR30) revealed changes not measured in the previous generation. Effects of the bifenthrin treatments were not observed until the F2 generation, which were exposed to the chemicals indirectly as germ cells. Our results indicate that effects of EDCs and their interactions with abiotic factors, may not be adequately represented by singular generation testing. These findings will contribute to the determination of the risk of EDC contamination to organisms inhabiting contaminated waterways under changing temperature regimes.
Collapse
Affiliation(s)
- Bethany M DeCourten
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, United States of America.,Department of Biology and Marine Biology, University of North Carolina at Wilmington, Wilmington, NC, United States of America
| | - Richard E Connon
- Department of Anatomy, Physiology and Cell Biology, University of California, Davis, CA, United States of America
| | - Susanne M Brander
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, United States of America.,Department of Biology and Marine Biology, University of North Carolina at Wilmington, Wilmington, NC, United States of America
| |
Collapse
|
19
|
Cabas I, Chaves-Pozo E, Mulero V, García-Ayala A. Role of estrogens in fish immunity with special emphasis on GPER1. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 89:102-110. [PMID: 30092317 DOI: 10.1016/j.dci.2018.08.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 08/03/2018] [Accepted: 08/03/2018] [Indexed: 06/08/2023]
Abstract
It is well accepted that estrogens, the primary female sex hormones, play a key role in modulating different aspects of the immune response. Moreover, estrogens have been linked with the sexual dimorphism observed in some immune disorders, such as chronic inflammatory and autoimmune diseases. Nevertheless, their effects are often controversial and depend on several factors, such as the pool of estrogen receptors (ERs) involved in the response. Their classical mode of action is through nuclear ERs, which act as transcription factors, promoting the regulation of target genes. However, it has long been noted that some of the estrogen-mediated effects cannot be explained by these classical receptors, since they are rapid and mediated by non-genomic signaling pathways. Hence, the interest in membrane ERs, especially in G protein-coupled estrogen receptor 1 (GPER1), has grown in recent years. Although the presence of nuclear ERs, and ER signaling, in immune cells in mammals and fish has been well documented, information on membrane ERs is much scarcer. In this context, the present manuscript aims to review our knowledge concerning the effect of estrogens on fish immunity, with special emphasis on GPER1. For example, the numerous tools developed over recent years allowed us to report for the first time that the regulation of fish granulocyte functions by estrogens through GPER1 predates the split of fish and tetrapods more than 450 million years ago, pointing to the relevance of estrogens as modulators of the immune responses, and the pivotal role of GPER1 in immunity.
Collapse
Affiliation(s)
- Isabel Cabas
- Department of Cell Biology and Histology, Faculty of Biology, University of Murcia, IMIB-Arrixaca, Murcia, Spain.
| | - Elena Chaves-Pozo
- Centro Oceanográfico de Murcia, Instituto Español de Oceanografía (IEO), Murcia, Spain
| | - Victoriano Mulero
- Department of Cell Biology and Histology, Faculty of Biology, University of Murcia, IMIB-Arrixaca, Murcia, Spain
| | - Alfonsa García-Ayala
- Department of Cell Biology and Histology, Faculty of Biology, University of Murcia, IMIB-Arrixaca, Murcia, Spain
| |
Collapse
|
20
|
Li J, Huang D, Sun X, Li X, Cheng CHK. Zinc mediates the action of androgen in acting as a downstream effector of luteinizing hormone on oocyte maturation in zebrafish†. Biol Reprod 2018; 100:468-478. [DOI: 10.1093/biolre/ioy224] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 08/01/2018] [Indexed: 12/18/2022] Open
Affiliation(s)
- Jianzhen Li
- College of Life Sciences, Northwest Normal University, Lanzhou, China
| | - Duo Huang
- School of Biomedical Sciences, The Chinese University of Hong Kong-Shandong University Joint Laboratory on Reproductive Genetics, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - Xiao Sun
- School of Biomedical Sciences, The Chinese University of Hong Kong-Shandong University Joint Laboratory on Reproductive Genetics, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - Xuehui Li
- College of Life Sciences, Northwest Normal University, Lanzhou, China
| | - Christopher H K Cheng
- School of Biomedical Sciences, The Chinese University of Hong Kong-Shandong University Joint Laboratory on Reproductive Genetics, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| |
Collapse
|
21
|
Effects of hypoxia exposure on apoptosis and expression of membrane steroid receptors, ZIP9, mPRα, and GPER in Atlantic croaker ovaries. Comp Biochem Physiol A Mol Integr Physiol 2018; 224:84-92. [DOI: 10.1016/j.cbpa.2018.07.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 07/03/2018] [Accepted: 07/03/2018] [Indexed: 11/17/2022]
|
22
|
Crowder CM, Romano SN, Gorelick DA. G Protein-Coupled Estrogen Receptor Is Not Required for Sex Determination or Ovary Function in Zebrafish. Endocrinology 2018; 159:3515-3523. [PMID: 30169775 PMCID: PMC6137278 DOI: 10.1210/en.2018-00685] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 08/17/2018] [Indexed: 01/22/2023]
Abstract
Estrogens regulate vertebrate development and function through binding to nuclear estrogen receptors α and β (ERα and ERβ) and the G protein-coupled estrogen receptor (GPER). Studies in mutant animal models demonstrated that ERα and ERβ are required for normal ovary development and function. However, the degree to which GPER signaling contributes to ovary development and function is less well understood. Previous studies using cultured fish oocytes found that estradiol inhibits oocyte maturation in a GPER-dependent manner, but whether GPER regulates oocyte maturation in vivo is not known. To test the hypothesis that GPER regulates oocyte maturation in vivo, we assayed ovary development and function in gper mutant zebrafish. We found that homozygous mutant gper embryos developed into male and female adults with normal sex ratios. Adult mutant fish exhibited normal secondary sex characteristics and fertility. Additionally, mutant ovaries were histologically normal. We observed no differences in the number of immature versus mature oocytes in mutant versus wild-type ovaries from both young and aged adults. Furthermore, expression of genes associated with sex determination and ovary function was normal in gper mutant ovaries compared with wild type. Our findings suggest that GPER is not required for sex determination, ovary development, or fertility in zebrafish.
Collapse
Affiliation(s)
- Camerron M Crowder
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Shannon N Romano
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Daniel A Gorelick
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Molecular and Cellular Biology, Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas
- Correspondence: Daniel A. Gorelick, PhD, Department of Molecular and Cellular Biology, Center for Precision Environmental Health, Baylor College of Medicine, Alkek Building, Suite N1317.02, One Baylor Plaza, BCM229, Houston, Texas 77030. E-mail:
| |
Collapse
|
23
|
Pang Y, Thomas P. Role of natriuretic peptide receptor 2-mediated signaling in meiotic arrest of zebrafish oocytes and its estrogen regulation through G protein-coupled estrogen receptor (Gper). Gen Comp Endocrinol 2018; 265:180-187. [PMID: 29574150 DOI: 10.1016/j.ygcen.2018.03.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 03/07/2018] [Accepted: 03/20/2018] [Indexed: 11/30/2022]
Abstract
Natriuretic peptide type C (NPPC) and its receptor, natriuretic peptide receptor 2 (NPR2), have essential roles in maintaining meiotic arrest of oocytes in several mammalian species. However, it is not known if a similar mechanism exists in non-mammalian vertebrates. Using zebrafish as a model, we show that Nppc is expressed in ovarian follicle cells, whereas Npr2 is mainly detected in oocytes. Treatment of intact and defolliculated oocytes with 100 nM NPPC for 6 h caused a large increase in cGMP concentrations, and a significant decrease in oocyte maturation (OM), an effect that was mimicked by treatment with 8-Br-cGMP. Treatment with E2 and G-1, the specific GPER agonist, also increased cGMP levels. Cyclic AMP levels were also increased by treatments with 8-Br-cGMP, E2 and G1. The estrogen upregulation of cAMP levels was blocked by co-treatment with AG1478, an inhibitor of EGFR activation. Gene expression of npr2, but not nppc, was significantly upregulated in intact oocytes by 6 h treatments with 20 nM E2 and G-1. Both cilostamide, a phosphodiesterase 3 (PDE3) inhibitor, and rolipram, a PDE4 inhibitor, significantly decreased OM of intact and defolliculated oocytes, and enhanced the inhibitory effects of E2 and G-1 on OM. These findings indicate the presence of a Nppc/Npr2/cGMP pathway maintaining meiotic arrest in zebrafish oocytes that is upregulated by estrogen activation of Gper. Collectively, the results suggest that Nppc through Npr2 cooperates with E2 through Gper in upregulation of cGMP levels to inhibit phosphodiesterase activity resulting in maintenance of oocyte meiotic arrest in zebrafish.
Collapse
Affiliation(s)
- Yefei Pang
- Marine Science Institute, University of Texas at Austin, 750 Channel View Drive, Port Aransas, TX 78373, USA.
| | - Peter Thomas
- Marine Science Institute, University of Texas at Austin, 750 Channel View Drive, Port Aransas, TX 78373, USA.
| |
Collapse
|
24
|
Differential expression of gonadotropin and estrogen receptors and oocyte cytology during follicular maturation associated with egg viability in European eel (Anguilla anguilla). Comp Biochem Physiol A Mol Integr Physiol 2018; 221:44-54. [DOI: 10.1016/j.cbpa.2018.03.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 03/15/2018] [Accepted: 03/15/2018] [Indexed: 11/21/2022]
|
25
|
Romano SN, Gorelick DA. Crosstalk between nuclear and G protein-coupled estrogen receptors. Gen Comp Endocrinol 2018; 261:190-197. [PMID: 28450143 PMCID: PMC5656538 DOI: 10.1016/j.ygcen.2017.04.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 04/04/2017] [Accepted: 04/22/2017] [Indexed: 10/19/2022]
Abstract
In 2005, two groups independently discovered that the G protein-coupled receptor GPR30 binds estradiol in cultured cells and, in response, initiates intracellular signaling cascades Revankar et al. (2005), Thomas et al. (2005). GPR30 is now referred to as GPER, the G-protein coupled estrogen receptor Prossnitz and Arterburn (2015). While studies in animal models are illuminating GPER function, there is controversy as to whether GPER acts as an autonomous estrogen receptor in vivo, or whether GPER interacts with nuclear estrogen receptor signaling pathways in response to estrogens. Here, we review the evidence that GPER acts as an autonomous estrogen receptor in vivo and discuss experimental approaches to test this hypothesis directly. We propose that the degree to which GPER influences nuclear estrogen receptor signaling likely depends on cell type, developmental stage and pathology.
Collapse
Affiliation(s)
- Shannon N Romano
- Department of Pharmacology & Toxicology, University of Alabama at Birmingham, USA
| | - Daniel A Gorelick
- Department of Pharmacology & Toxicology, University of Alabama at Birmingham, USA.
| |
Collapse
|
26
|
Zhu D, Yang L, Huang J, Zhou F, Yang Q, Jiang S, Jiang S. The comprehensive expression analysis of the G protein-coupled receptor from Penaeus monodon indicating it participates in innate immunity and anti-ammonia nitrogen stress. FISH & SHELLFISH IMMUNOLOGY 2018; 75:17-26. [PMID: 29410275 DOI: 10.1016/j.fsi.2018.01.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 01/09/2018] [Accepted: 01/12/2018] [Indexed: 06/07/2023]
Abstract
The G protein-coupled receptors (GPCRs) composed a superfamily that played an important role in physiological processes of crustaceans, with multiple functions such as growth and development, acting as a defense against stimulations from external factors. In this paper, one kind of GPCRs were identified from Penaeus monodon, called PmGPCR, included an open reading frame (ORF) of 1113 bp. Bioinformatic analysis showed that PmGPCR protein had the typical structure of seven transmembrane domains (7TM), especially the special Asp-Arg-Try motif (DRY motif) between the third transmembrane structures (TM3) and the second intracellular loops (IL-2) which can prove that PmGPCR belongs to the rhodopsin-like family. The analyses of phylogenetic tree indicated that the amino acid sequence of PmGPCR should be merged into Procambarus clarkiic with high identity (98%). Quantitative real-time PCR (q RT-PCR) revealed that PmGPCR mRNA was highly expressed in hepatopancreas, abdominal ganglia and lymph, in which it was significantly higher than that of other tissues (P < 0.05). In addition, the expression of PmGPCR was analyzed during three days post-stimulation with the gram-positive/negative bacteria, the mRNA expression level increased after challenged with gram - positive bacteria in hepatopancreas, lymph and intestines. During the development stages, PmGPCR showed significantly higher expression in nauplius, zoea III, mysis III and post larvae stages than that in other development stages. Meanwhile, the highest transcripts expression of PmGPCR in abdominal ganglia, hepatopancreas, lymph and intestines respectively appeared at D0, D1, D2 and D3/D4 stages of molting. High or low concentration of ammonia nitrogen up-regulated the expression level of PmGPCR at the initial stage in hepatopancreas and gill, and then down-regulated at 48 h. These results indicated PmGPCR may mediate the pathways that involved in growth and development process, survival in the adversity, in addition, provided the useful data to research GPCR-mediated physiological and biological process and explain the mechanisms to defense pathogens and anti-stress in shrimp.
Collapse
Affiliation(s)
- Dandan Zhu
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture, Guangzhou, 510300, PR China; College of Aqua-life Science and Technology, Shanghai Ocean University, Shanghai, PR China
| | - Lishi Yang
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture, Guangzhou, 510300, PR China
| | - Jianhua Huang
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture, Guangzhou, 510300, PR China; Shenzhen Base of South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shenzhen, 518108, PR China
| | - Falin Zhou
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture, Guangzhou, 510300, PR China
| | - Qibin Yang
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture, Guangzhou, 510300, PR China
| | - Song Jiang
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture, Guangzhou, 510300, PR China
| | - Shigui Jiang
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture, Guangzhou, 510300, PR China; Shenzhen Base of South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shenzhen, 518108, PR China.
| |
Collapse
|
27
|
Pérez C, Araneda C, Estay F, Díaz NF, Vizziano-Cantonnet D. Sex hormone-binding globulin b expression in the rainbow trout ovary prior to sex differentiation. Gen Comp Endocrinol 2018; 259:165-175. [PMID: 29180105 DOI: 10.1016/j.ygcen.2017.11.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 11/23/2017] [Accepted: 11/23/2017] [Indexed: 10/18/2022]
Abstract
Salmonids have two sex hormone-binding globulin (Shbg) paralogs. Shbga is mainly expressed in the liver, while Shbgb is secreted by the granulosa cells of the rainbow trout ovary. Coexpression of shbgb and the gonadal aromatase cyp19a1a mRNAs been observed in granulosa cells, suggesting a physiological coordination between Shbgb expression and estrogen synthesis. As estrogens are essential for female sex determination in the fish ovary, we propose that Shbgb participates in early ovarian differentiation, either by binding with estrogen or through another mechanism that remains to be discovered. To elucidate this potential role, monosex populations of female trout were studied during the molecular ovarian differentiation period (28-56 dpf). shbgb mRNA expression was measured using qPCR and compared with expression of genes for other ovarian markers (cyp19a1a, foxl2, follistatin, and estrogen receptors). shbgb transcript expression was detected during the final stages of embryonic development (21-26 dpf) and during molecular ovarian differentiation (32-52 dpf) after hatching (which occurred at 31 dpf). In situ hybridization localized shbgb transcription to the undifferentiated ovary at 42 dpf, and shbgb and cyp19a1a mRNA showed similar expression patterns. These results suggest that Shbgb is involved in early ovarian differentiation, supporting an important role for the salmonid shbgb gene in sex determination.
Collapse
Affiliation(s)
- Claudio Pérez
- Laboratorio de Genética y Biotecnología en Acuicultura, Facultad de Ciencias Agronómicas, Universidad de Chile, Avenida Santa Rosa #11315, Santiago de, Chile; Programa Cooperativo de Doctorado en Acuicultura, Escuela de Postgrado, Facultad de Ciencias Agronómicas, Universidad de Chile, Avenida Santa Rosa #11315, Santiago de, Chile
| | - Cristian Araneda
- Laboratorio de Genética y Biotecnología en Acuicultura, Facultad de Ciencias Agronómicas, Universidad de Chile, Avenida Santa Rosa #11315, Santiago de, Chile.
| | - Francisco Estay
- Piscicultura Huililco Ltda, Camino a Caburgua km 17, Pucón IX Región, Chile
| | - Nelson F Díaz
- Laboratorio de Genética y Biotecnología en Acuicultura, Facultad de Ciencias Agronómicas, Universidad de Chile, Avenida Santa Rosa #11315, Santiago de, Chile
| | - Denise Vizziano-Cantonnet
- Laboratorio de Fisiología de la Reproducción y Ecología de Peces, Facultad de Ciencias, Universidad de la República Oriental del Uruguay, Iguá 4225, Montevideo 11400, Uruguay
| |
Collapse
|
28
|
Pinto PIS, Andrade AR, Estêvão MD, Alvarado MV, Felip A, Power DM. Duplicated membrane estrogen receptors in the European sea bass (Dicentrarchus labrax): Phylogeny, expression and regulation throughout the reproductive cycle. J Steroid Biochem Mol Biol 2018; 178:234-242. [PMID: 29288793 DOI: 10.1016/j.jsbmb.2017.12.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 12/22/2017] [Accepted: 12/23/2017] [Indexed: 10/18/2022]
Abstract
The numerous estrogen functions reported across vertebrates have been classically explained by their binding to specific transcription factors, the nuclear estrogen receptors (ERs). Rapid non-genomic estrogenic responses have also been recently identified in vertebrates including fish, which can be mediated by membrane receptors such as the G protein-coupled estrogen receptor (Gper). In this study, two genes for Gper, namely gpera and gperb, were identified in the genome of a teleost fish, the European sea bass. Phylogenetic analysis indicated they were most likely retained after the 3R teleost-specific whole genome duplication and raises questions about their function in male and female sea bass. Gpera expression was mainly restricted to brain and pituitary in both sexes while gperb had a widespread tissue distribution with higher expression levels in gill filaments, kidney and head kidney. Both receptors were detected in the hypothalamus and pituitary of both sexes and significant changes in gpers expression were observed throughout the annual reproductive season. In female pituitaries, gpera showed an overall increase in expression throughout the reproductive season while gperb levels remained constant. In the hypothalamus, gpera had a higher expression during vitellogenesis and decreased in fish entering the ovary maturation and ovulation stage, while gperb expression increased at the final atresia stage. In males, gpers expression was constant in the hypothalamus and pituitary throughout the reproductive cycle apart from the mid- to late testicular development stage transition when a significant up-regulation of gpera occurred in the pituitary. The differential sex, seasonal and subtype-specific expression patterns detected for the two novel gper genes in sea bass suggests they may have acquired different and/or complementary roles in mediating estrogens actions in fish, namely on the neuroendocrine control of reproduction.
Collapse
Affiliation(s)
| | | | - M Dulce Estêvão
- CCMAR - Centre of Marine Sciences, Faro, Portugal; Escola Superior de Saúde, Universidade do Algarve, Av. Dr. Adelino da Palma Carlos, 8000-510 Faro, Portugal.
| | - M Victoria Alvarado
- CCMAR - Centre of Marine Sciences, Faro, Portugal; Institute of Aquaculture Torre de la Sal (IATS-CSIC), Castellón, Spain.
| | - Alicia Felip
- Institute of Aquaculture Torre de la Sal (IATS-CSIC), Castellón, Spain.
| | | |
Collapse
|
29
|
Zheng H, Li H, Tan W, Xu C, Jia L, Wang D, Li Z, Sun G, Kang X, Yan F, Liu X. Oestrogen regulates the expression of cathepsin E-A-like gene through ERΒ in liver of chicken (Gallus gallus). J Genet 2018; 97:145-155. [PMID: 29666334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The cathepsin E-A-like, also known as 'similar to nothepsin', is a new member of the aspartic protease family, which may take part in processing of egg yolk macromolecules, due to it was identified in the chicken egg-yolk. Previously, studies have suggested that the expression of cathepsin E-A-like increased gradually during sexual maturation of pullets, but the exact regulation mechanism is poorly understood. In this study, to gain insight into the function and regulation mechanism of the gene in egg-laying hen, we cloned the cathepsin E-A-like gene and evaluated its evolutionary origin by using both phylogenetic and syntenic methods. The mode of the gene expression regulation was analysed through stimulating juvenile hens with 17β-estradiol and chicken embryo hepatocytes with 17β-estradiol combined with oestrogen receptor antagonists including MPP, ICI 182,780 and tamoxifen. Our results showed that cathepsin E-A-like was an orthologoues gene with nothepsin, which is present in birds but not in mammals. The expression of cathepsin E-A-like significantly increased in a dose-dependent manner after the juvenile hens were treated with 17β-estradiol (P < 0.05). Compared with the 17β-estradiol treatment group, the expression of cathepsin E-A-like was not significantly changed when the hepatocytes were treated with 17β-estradiol combined with MPP (P < 0.05). In contrast, compared with the 17β-estradiol combined with MPP treatment group, the expression of cathepsin E-A-like was significantly downregulated when the hepatocytes were treated with 17β-estradiol combined with tamoxifen or ICI 182,780 (P < 0.05). These results demonstrated that cathepsin E-A-like shared the same evolutionary origin with nothepsin. The expression of cathepsin E-A-like was regulated by oestrogen, and the regulative effect was predominantly mediated through ER-Β in liver of chicken.
Collapse
Affiliation(s)
- Hang Zheng
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, People's Republic of China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Oestrogen regulates the expression of cathepsin E-A-like gene through ER
$$\upbeta $$
β
in liver of chicken (Gallus gallus). J Genet 2018. [DOI: 10.1007/s12041-018-0890-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
31
|
Diotel N, Charlier TD, Lefebvre d'Hellencourt C, Couret D, Trudeau VL, Nicolau JC, Meilhac O, Kah O, Pellegrini E. Steroid Transport, Local Synthesis, and Signaling within the Brain: Roles in Neurogenesis, Neuroprotection, and Sexual Behaviors. Front Neurosci 2018; 12:84. [PMID: 29515356 PMCID: PMC5826223 DOI: 10.3389/fnins.2018.00084] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 02/02/2018] [Indexed: 01/18/2023] Open
Abstract
Sex steroid hormones are synthesized from cholesterol and exert pleiotropic effects notably in the central nervous system. Pioneering studies from Baulieu and colleagues have suggested that steroids are also locally-synthesized in the brain. Such steroids, called neurosteroids, can rapidly modulate neuronal excitability and functions, brain plasticity, and behavior. Accumulating data obtained on a wide variety of species demonstrate that neurosteroidogenesis is an evolutionary conserved feature across fish, birds, and mammals. In this review, we will first document neurosteroidogenesis and steroid signaling for estrogens, progestagens, and androgens in the brain of teleost fish, birds, and mammals. We will next consider the effects of sex steroids in homeostatic and regenerative neurogenesis, in neuroprotection, and in sexual behaviors. In a last part, we will discuss the transport of steroids and lipoproteins from the periphery within the brain (and vice-versa) and document their effects on the blood-brain barrier (BBB) permeability and on neuroprotection. We will emphasize the potential interaction between lipoproteins and sex steroids, addressing the beneficial effects of steroids and lipoproteins, particularly HDL-cholesterol, against the breakdown of the BBB reported to occur during brain ischemic stroke. We will consequently highlight the potential anti-inflammatory, anti-oxidant, and neuroprotective properties of sex steroid and lipoproteins, these latest improving cholesterol and steroid ester transport within the brain after insults.
Collapse
Affiliation(s)
- Nicolas Diotel
- Université de La Réunion, Institut National de la Santé et de la Recherche Médicale, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien, Saint-Denis de La Réunion, France
| | - Thierry D. Charlier
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| | - Christian Lefebvre d'Hellencourt
- Université de La Réunion, Institut National de la Santé et de la Recherche Médicale, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien, Saint-Denis de La Réunion, France
| | - David Couret
- Université de La Réunion, Institut National de la Santé et de la Recherche Médicale, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien, Saint-Denis de La Réunion, France
- CHU de La Réunion, Saint-Denis, France
| | | | - Joel C. Nicolau
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| | - Olivier Meilhac
- Université de La Réunion, Institut National de la Santé et de la Recherche Médicale, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien, Saint-Denis de La Réunion, France
- CHU de La Réunion, Saint-Denis, France
| | - Olivier Kah
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| | - Elisabeth Pellegrini
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| |
Collapse
|
32
|
Garcia-Reyero N, Jayasinghe BS, Kroll KJ, Sabo-Attwood T, Denslow ND. Estrogen signaling through both membrane and nuclear receptors in the liver of fathead minnow. Gen Comp Endocrinol 2018; 257:50-66. [PMID: 28733229 DOI: 10.1016/j.ygcen.2017.07.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 05/04/2017] [Accepted: 07/16/2017] [Indexed: 11/24/2022]
Abstract
Estradiol is a potent sex steroid hormone that controls reproduction and other cellular pathways in fish. It is known to regulate important proteins such as vitellogenin, the egg yolk precursor protein, and zona radiata proteins that form the eggshell for fish eggs. These proteins are made in the liver and transported out into the blood from where they are taken up into the ovary during oogenesis. Estradiol can exert its influence directly through soluble nuclear receptors (there are three in fish) or indirectly through membrane receptors and a phosphorylation cascade. Often there is coordination through both genomic and non-genomic pathways. We have used a toxicogenomics approach to determine the contribution of genomic and non-genomic regulation in the liver of fathead minnows exposed to 5ng ethinylestradiol per liter or to a mixture of 5ng ethinylestradiol and 100ng ZM189,154 (ZM) per liter. ZM has previously been shown to be a "perfect" antagonist for the fish nuclear estrogen receptors but has displayed agonistic activities for membrane receptors. We find that both nuclear and membrane receptors contribute to the biosynthesis of vitellogenin 1 and estrogen receptor one (Esr1), among others. In addition, lipid metabolism pathways appear to require both activities.
Collapse
Affiliation(s)
- Natàlia Garcia-Reyero
- Environmental Laboratory, US Army Engineer Research & Development Center, Vicksburg, MS 39180, USA; Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32611, USA.
| | - B Sumith Jayasinghe
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32611, USA
| | - Kevin J Kroll
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32611, USA
| | - Tara Sabo-Attwood
- Department of Environmental and Global Health, University of Florida, Gainesville, FL 32611, USA
| | - Nancy D Denslow
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
33
|
Barton M, Filardo EJ, Lolait SJ, Thomas P, Maggiolini M, Prossnitz ER. Twenty years of the G protein-coupled estrogen receptor GPER: Historical and personal perspectives. J Steroid Biochem Mol Biol 2018; 176:4-15. [PMID: 28347854 PMCID: PMC5716468 DOI: 10.1016/j.jsbmb.2017.03.021] [Citation(s) in RCA: 157] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/22/2017] [Accepted: 03/23/2017] [Indexed: 12/24/2022]
Abstract
Estrogens play a critical role in many aspects of physiology, particularly female reproductive function, but also in pathophysiology, and are associated with protection from numerous diseases in premenopausal women. Steroids and the effects of estrogen have been known for ∼90 years, with the first evidence for a receptor for estrogen presented ∼50 years ago. The original ancestral steroid receptor, extending back into evolution more than 500 million years, was likely an estrogen receptor, whereas G protein-coupled receptors (GPCRs) trace their origins back into history more than one billion years. The classical estrogen receptors (ERα and ERβ) are ligand-activated transcription factors that confer estrogen sensitivity upon many genes. It was soon apparent that these, or novel receptors may also be responsible for the "rapid"/"non-genomic" membrane-associated effects of estrogen. The identification of an orphan GPCR (GPR30, published in 1996) opened a new field of research with the description in 2000 that GPR30 expression is required for rapid estrogen signaling. In 2005-2006, the field was greatly stimulated by two studies that described the binding of estrogen to GPR30-expressing cell membranes, followed by the identification of a GPR30-selective agonist (that lacked binding and activity towards ERα and ERβ). Renamed GPER (G protein-coupled estrogen receptor) by IUPHAR in 2007, the total number of articles in PubMed related to this receptor recently surpassed 1000. In this article, the authors present personal perspectives on how they became involved in the discovery and/or advancement of GPER research. These areas include non-genomic effects on vascular tone, receptor cloning, molecular and cellular biology, signal transduction mechanisms and pharmacology of GPER, highlighting the roles of GPER and GPER-selective compounds in diseases such as obesity, diabetes, and cancer and the obligatory role of GPER in propagating cardiovascular aging, arterial hypertension and heart failure through the stimulation of Nox expression.
Collapse
Affiliation(s)
- Matthias Barton
- Molecular Internal Medicine, University of Zürich, 8057 Zürich, Switzerland.
| | - Edward J Filardo
- Rhode Island Hospital, Brown University, Providence, RI 02903, USA
| | - Stephen J Lolait
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, University of Bristol, Bristol, UK
| | - Peter Thomas
- Marine Science Institute, University of Texas at Austin, Port Aransas, TX 78373, USA
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Eric R Prossnitz
- Department of Internal Medicine, University of New Mexico Health Sciences Center and University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA.
| |
Collapse
|
34
|
Thomas P. Reprint of "Role of G protein-coupled estrogen receptor (GPER/GPR30) in maintenance of meiotic arrest in fish oocytes". J Steroid Biochem Mol Biol 2018; 176:23-30. [PMID: 29102625 DOI: 10.1016/j.jsbmb.2017.10.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 11/23/2016] [Accepted: 12/16/2016] [Indexed: 01/07/2023]
Abstract
An essential role for GPER (formerly known as GPR30) in regulating mammalian reproduction has not been identified to date, although it has shown to be involved in the regulation a broad range of other estrogen-dependent functions. In contrast, an important reproductive role for GPER in the maintenance of oocyte meiotic arrest has been identified in teleost fishes, which is briefly reviewed here. Recent studies have clearly shown that ovarian follicle production of estradiol-17β (E2) maintains meiotic arrest in several teleost species through activation of GPER coupled to a stimulatory G protein (Gs) on oocyte plasma membranes, resulting in stimulation of cAMP production and maintenance of elevated cAMP levels. Studies with denuded zebrafish oocytes and with microinjection of GPER antisense oligonucleotides into oocytes have demonstrated the requirement for both ovarian follicle production of estrogens and expression of GPER on the oocyte surface for maintenance of meiotic arrest. This inhibitory action of E2 on the resumption of meiosis is mimicked by the GPER-selective agonist G-1, by the GPER agonists and nuclear ER antagonists, ICI 182,780 and tamoxifen, and also by the xenoestrogen bisphenol-A (BPA) and related alkylphenols. GPER also maintains meiotic arrest of zebrafish oocytes through estrogen- and BPA-dependent GPER activation of epidermal growth factor receptor (EGFR) and mitogen-activated protein kinase (MAPK) signaling. Interestingly, progesterone receptor component 1 (PGRMC1) is also involved in estrogen maintenance of meiotic arrest through regulation of EGFR expression on the oocyte plasma membrane. The preovulatory surge in LH secretion induces the ovarian synthesis of progestin hormones that activate a membrane progestin receptor alpha (mPRα)/inhibitory G protein (Gi) pathway. It also increases ovarian synthesis of the catecholestrogen, 2-hydroxy-estradiol-17β (2-OHE2) which inhibits the GPER/Gs/adenylyl cyclase pathway. Both of these LH actions cause declines in oocyte cAMP levels resulting in the resumption of meiosis. GPER is also present on murine oocytes but there are no reports of studies investigating its possible involvement in maintaining meiotic arrest in mammals.
Collapse
Affiliation(s)
- Peter Thomas
- Marine Science Institute, University of Texas at Austin, 750 Channel View Drive, Port Aransas, TX 78373, United States.
| |
Collapse
|
35
|
Romano SN, Edwards HE, Souder JP, Ryan KJ, Cui X, Gorelick DA. G protein-coupled estrogen receptor regulates embryonic heart rate in zebrafish. PLoS Genet 2017; 13:e1007069. [PMID: 29065151 PMCID: PMC5669493 DOI: 10.1371/journal.pgen.1007069] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 11/03/2017] [Accepted: 10/11/2017] [Indexed: 01/31/2023] Open
Abstract
Estrogens act by binding to estrogen receptors alpha and beta (ERα, ERβ), ligand-dependent transcription factors that play crucial roles in sex differentiation, tumor growth and cardiovascular physiology. Estrogens also activate the G protein-coupled estrogen receptor (GPER), however the function of GPER in vivo is less well understood. Here we find that GPER is required for normal heart rate in zebrafish embryos. Acute exposure to estrogens increased heart rate in wildtype and in ERα and ERβ mutant embryos but not in GPER mutants. GPER mutant embryos exhibited reduced basal heart rate, while heart rate was normal in ERα and ERβ mutants. We detected gper transcript in discrete regions of the brain and pituitary but not in the heart, suggesting that GPER acts centrally to regulate heart rate. In the pituitary, we observed gper expression in cells that regulate levels of thyroid hormone triiodothyronine (T3), a hormone known to increase heart rate. Compared to wild type, GPER mutants had reduced levels of T3 and estrogens, suggesting pituitary abnormalities. Exposure to exogenous T3, but not estradiol, rescued the reduced heart rate phenotype in gper mutant embryos, demonstrating that T3 acts downstream of GPER to regulate heart rate. Using genetic and mass spectrometry approaches, we find that GPER regulates maternal estrogen levels, which are required for normal embryonic heart rate. Our results demonstrate that estradiol plays a previously unappreciated role in the acute modulation of heart rate during zebrafish embryonic development and suggest that GPER regulates embryonic heart rate by altering maternal estrogen levels and embryonic T3 levels. Estrogen hormones are important for the formation and function of the nervous, reproductive and cardiovascular systems. Here we report that acute exposure to estrogens increases heart rate, a previously unappreciated function of estrogens. Using zebrafish with mutations in genes that respond to estrogens, we found that heart rate is regulated not by the typical molecules that respond to estrogens–the nuclear estrogen receptors–but rather by a different molecule, the G protein-coupled estrogen receptor. We also show that estrogens increase heart rate by increasing levels of thyroid hormone. Our results reveal a new function for the G protein-coupled estrogen receptor and a new connection between estrogens and thyroid hormone. Environmental compounds that mimic estrogens can be harmful because they can influence gonad function. Our results suggest that endocrine disrupting compounds may also influence cardiac function.
Collapse
Affiliation(s)
- Shannon N. Romano
- Department of Pharmacology & Toxicology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Hailey E. Edwards
- Department of Pharmacology & Toxicology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Jaclyn Paige Souder
- Department of Pharmacology & Toxicology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Kevin J. Ryan
- Department of Pharmacology & Toxicology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Xiangqin Cui
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Daniel A. Gorelick
- Department of Pharmacology & Toxicology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- * E-mail:
| |
Collapse
|
36
|
Lu H, Cui Y, Jiang L, Ge W. Functional Analysis of Nuclear Estrogen Receptors in Zebrafish Reproduction by Genome Editing Approach. Endocrinology 2017; 158:2292-2308. [PMID: 28398516 DOI: 10.1210/en.2017-00215] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 04/03/2017] [Indexed: 01/09/2023]
Abstract
Estrogens signal through both nuclear and membrane receptors with most reported effects being mediated via the nuclear estrogen receptors (nERs). Although much work has been reported on nERs in the zebrafish, there is a lack of direct genetic evidence for their functional roles and importance in reproduction. To address this issue, we undertook this study to disrupt all three nERs in the zebrafish, namely esr1 (ERα), esr2a (ERβII), and esr2b (ERβI), by the genome-editing technology clustered regularly interspaced short palindromic repeats and its associated nuclease (CRISPR/Cas9). Using this loss-of-function genetic approach, we successfully created three mutant zebrafish lines with each nER knocked out. In addition, we also generated all possible double and triple knockouts of the three nERs. The phenotypes of these mutants in reproduction were analyzed in all single, double, and triple nER knockouts in both females and males. Surprisingly, all three single nER mutant fish lines display normal reproductive development and function in both females and males, suggesting functional redundancy among these nERs. Further analysis of double and triple knockouts showed that nERs, especially Esr2a and Esr2b, were essential for female reproduction, and loss of these two nERs led to an arrest of folliculogenesis at previtellogenic stage II followed by sex reversal from female to male. In addition, the current study also revealed a unique role for Esr2a in follicle cell proliferation and transdifferentiation, follicle growth, and chorion formation. Taken together, this study provides the most comprehensive genetic analysis for differential functions of esr1, esr2a, and esr2b in fish reproduction.
Collapse
Affiliation(s)
- Huijie Lu
- Centre of Reproduction, Development, and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Yong Cui
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Liwen Jiang
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Wei Ge
- Centre of Reproduction, Development, and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| |
Collapse
|
37
|
Thomas P. Role of G-protein-coupled estrogen receptor (GPER/GPR30) in maintenance of meiotic arrest in fish oocytes. J Steroid Biochem Mol Biol 2017; 167:153-161. [PMID: 28007532 DOI: 10.1016/j.jsbmb.2016.12.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 11/23/2016] [Accepted: 12/16/2016] [Indexed: 02/08/2023]
Abstract
An essential role for GPER (formerly known as GPR30) in regulating mammalian reproduction has not been identified to date, although it has shown to be involved in the regulation a broad range of other estrogen-dependent functions. In contrast, an important reproductive role for GPER in the maintenance of oocyte meiotic arrest has been identified in teleost fishes, which is briefly reviewed here. Recent studies have clearly shown that ovarian follicle production of estradiol-17β (E2) maintains meiotic arrest in several teleost species through activation of GPER coupled to a stimulatory G protein (Gs) on oocyte plasma membranes resulting in stimulation of cAMP production and maintenance of elevated cAMP levels. Studies with denuded zebrafish oocytes and with microinjection of GPER antisense oligonucleotides into oocytes have demonstrated the requirement for both ovarian follicle production of estrogens and expression of GPER on the oocyte surface for maintenance of meiotic arrest. This inhibitory action of E2 on the resumption of meiosis is mimicked by the GPER-selective agonist G-1, by the GPER agonists and nuclear ER antagonists, ICI 182,780 and tamoxifen, and also by the xenoestrogen bisphenol-A (BPA) and related alkylphenols. GPER also maintains meiotic arrest of zebrafish oocytes through estrogen- and BPA-dependent GPER activation of epidermal growth factor receptor (EGFR) and mitogen-activated protein kinase (MAPK) signaling. Interestingly, progesterone receptor component 1 (PGRMC1) is also involved in estrogen maintenance of meiotic arrest through regulation of EGFR expression on the oocyte plasma membrane. The preovulatory surge in LH secretion induces the ovarian synthesis of progestin hormones that activate a membrane progestin receptor alpha (mPRα)/inhibitory G protein (Gi) pathway. It also increases ovarian synthesis of the catecholestrogen, 2-hydroxy-estradiol-17β (2-OHE2) which inhibits the GPER/Gs/adenylyl cyclase pathway. Both of these LH actions cause declines in oocyte cAMP levels resulting in the resumption of meiosis. GPER is also present on murine oocytes but there are no reports of studies investigating its possible involvement in maintaining meiotic arrest in mammals.
Collapse
Affiliation(s)
- Peter Thomas
- Marine Science Institute, University of Texas at Austin, 750 Channel View Drive, Port Aransas, TX 78373, United States.
| |
Collapse
|
38
|
Chen SX, Yang XZ, Deng Y, Huang J, Li Y, Sun Q, Yu CP, Zhu Y, Hong WS. Silver nanoparticles induce oocyte maturation in zebrafish (Danio rerio). CHEMOSPHERE 2017; 170:51-60. [PMID: 27974271 PMCID: PMC5575830 DOI: 10.1016/j.chemosphere.2016.12.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 11/21/2016] [Accepted: 12/04/2016] [Indexed: 05/18/2023]
Abstract
Public concern regarding silver nanoparticles (AgNPs) in the environment has been increasing since they can cause adverse effects in some aquatic species. However, few data are actually available on the effects of AgNPs on the germ cells. In the present study, we used the zebrafish ovarian follicle as a model to assess the potentially adverse effects of AgNPs on oocyte maturation (germinal vesicle breakdown, GVBD) in vitro. Similar to the maturation inducing hormone (17α, 20β-dihydroxy-4-pregnen-3-one), AgNPs induced GVBD, and reduced the total cyclic adenosine monophosphate (cAMP) concentration in zebrafish ovarian follicles. The results from transmission electron microscope observation and Hoechst 33342 staining clearly indicated that AgNPs induced apoptosis in ovarian follicle cells surrounding the oocyte. Similar to AgNPs, AgNO3 also induced GVBD, decreased cAMP concentration and induced apoptosis of ovarian follicle cells. However, the results from gene expression analysis showed that transcript levels of oxidative stress related genes were more sensitive to AgNPs than AgNO3. Further more, H2O2 has an ability to induce zebrafish oocytes maturation by induction of apoptosis in ovarian follicle cells. Taken together, the results from our study indicated that oxidative stress appeared to be one of important mechanisms in AgNP induced apoptosis in ovarian follicle cells, which further triggered the GVBD.
Collapse
Affiliation(s)
- Shi Xi Chen
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, PR China; Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, Xiamen University, Xiamen, 361102, PR China; State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, Xiamen University, Xiamen, 361102, PR China.
| | - Xiao Zhen Yang
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, PR China
| | - Ying Deng
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, PR China
| | - Jing Huang
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, PR China
| | - Yan Li
- CAS Key Laboratory of Urban Pollutant Conversion, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, 361021, PR China
| | - Qian Sun
- CAS Key Laboratory of Urban Pollutant Conversion, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, 361021, PR China
| | - Chang-Ping Yu
- CAS Key Laboratory of Urban Pollutant Conversion, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, 361021, PR China
| | - Yong Zhu
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, PR China; Department of Biology, East Carolina University, Greenville, NC, USA
| | - Wan Shu Hong
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, PR China; Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, Xiamen University, Xiamen, 361102, PR China
| |
Collapse
|
39
|
Szwejser E, Maciuszek M, Casanova-Nakayama A, Segner H, Verburg-van Kemenade BML, Chadzinska M. A role for multiple estrogen receptors in immune regulation of common carp. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 66:61-72. [PMID: 27062969 DOI: 10.1016/j.dci.2016.04.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 04/05/2016] [Accepted: 04/05/2016] [Indexed: 05/02/2023]
Abstract
Estrogens are important for bi-directional neuroendocrine-immune interaction. They act via nuclear estrogen receptors (ERα and ERβ) and/or G-protein coupled receptor - GPR30. We found expression of ERα, ERβ and GPR30 in carp lymphoid tissues and head kidney monocytes/macrophages, neutrophils and lymphocytes. Interestingly, ERβ is also expressed in some head kidney lymphocytes but not in naive PBLs. Immune stimulation altered the cell type specific profile of expression of these receptors, which depends on both activation and maturation stage. This implies direct leukocyte responsiveness to estrogen stimulation and therefore in vitro effects of 17β-estradiol (E2) on reactive oxygen species (ROS) production in monocytes/macrophages were determined. Short-time incubation with E2 increased ROS production in PMA-stimulated cells. Results comply with mediation by GPR30, partially functioning via phosphoinositide 3-kinase activation. These results furthermore demonstrate that neuroendocrine-immune communication via estrogen receptors is evolutionary conserved.
Collapse
Affiliation(s)
- Ewa Szwejser
- Department of Evolutionary Immunology, Institute of Zoology, Jagiellonian University, Gronostajowa 9, PL30-387 Krakow, Poland
| | - Magdalena Maciuszek
- Department of Evolutionary Immunology, Institute of Zoology, Jagiellonian University, Gronostajowa 9, PL30-387 Krakow, Poland
| | - Ayako Casanova-Nakayama
- Centre for Fish and Wildlife Health, University of Bern, Länggassstrasse 122, CH-3012 Bern, Switzerland
| | - Helmut Segner
- Centre for Fish and Wildlife Health, University of Bern, Länggassstrasse 122, CH-3012 Bern, Switzerland
| | - B M Lidy Verburg-van Kemenade
- Cell Biology and Immunology Group, Dept of Animal Sciences, Wageningen University, P.O. Box 338, 6700 AH Wageningen, The Netherlands
| | - Magdalena Chadzinska
- Department of Evolutionary Immunology, Institute of Zoology, Jagiellonian University, Gronostajowa 9, PL30-387 Krakow, Poland.
| |
Collapse
|
40
|
Morini M, Peñaranda DS, Vílchez MC, Tveiten H, Lafont AG, Dufour S, Pérez L, Asturiano JF. The expression of nuclear and membrane estrogen receptors in the European eel throughout spermatogenesis. Comp Biochem Physiol A Mol Integr Physiol 2017; 203:91-99. [DOI: 10.1016/j.cbpa.2016.08.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 08/03/2016] [Accepted: 08/24/2016] [Indexed: 02/02/2023]
|
41
|
Kwakowsky A, Milne MR, Waldvogel HJ, Faull RL. Effect of Estradiol on Neurotrophin Receptors in Basal Forebrain Cholinergic Neurons: Relevance for Alzheimer's Disease. Int J Mol Sci 2016; 17:E2122. [PMID: 27999310 PMCID: PMC5187922 DOI: 10.3390/ijms17122122] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 12/05/2016] [Accepted: 12/12/2016] [Indexed: 02/06/2023] Open
Abstract
The basal forebrain is home to the largest population of cholinergic neurons in the brain. These neurons are involved in a number of cognitive functions including attention, learning and memory. Basal forebrain cholinergic neurons (BFCNs) are particularly vulnerable in a number of neurological diseases with the most notable being Alzheimer's disease, with evidence for a link between decreasing cholinergic markers and the degree of cognitive impairment. The neurotrophin growth factor system is present on these BFCNs and has been shown to promote survival and differentiation on these neurons. Clinical and animal model studies have demonstrated the neuroprotective effects of 17β-estradiol (E2) on neurodegeneration in BFCNs. It is believed that E2 interacts with neurotrophin signaling on cholinergic neurons to mediate these beneficial effects. Evidence presented in our recent study confirms that altering the levels of circulating E2 levels via ovariectomy and E2 replacement significantly affects the expression of the neurotrophin receptors on BFCN. However, we also showed that E2 differentially regulates neurotrophin receptor expression on BFCNs with effects depending on neurotrophin receptor type and neuroanatomical location. In this review, we aim to survey the current literature to understand the influence of E2 on the neurotrophin system, and the receptors and signaling pathways it mediates on BFCN. In addition, we summarize the physiological and pathophysiological significance of E2 actions on the neurotrophin system in BFCN, especially focusing on changes related to Alzheimer's disease.
Collapse
Affiliation(s)
- Andrea Kwakowsky
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand.
| | - Michael R Milne
- School of Biomedical Sciences, Queensland Brain Institute, Clem Jones Centre for Ageing Dementia Research, The University of Queensland, Brisbane 4072, QLD, Australia.
| | - Henry J Waldvogel
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand.
| | - Richard L Faull
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand.
| |
Collapse
|
42
|
Lafont AG, Rousseau K, Tomkiewicz J, Dufour S. Three nuclear and two membrane estrogen receptors in basal teleosts, Anguilla sp.: Identification, evolutionary history and differential expression regulation. Gen Comp Endocrinol 2016; 235:177-191. [PMID: 26654744 DOI: 10.1016/j.ygcen.2015.11.021] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 11/26/2015] [Accepted: 11/29/2015] [Indexed: 12/17/2022]
Abstract
Estrogens interact with classical intracellular nuclear receptors (ESR), and with G-coupled membrane receptors (GPER). In the eel, we identified three nuclear (ESR1, ESR2a, ESR2b) and two membrane (GPERa, GPERb) estrogen receptors. Duplicated ESR2 and GPER were also retrieved in most extant teleosts. Phylogeny and synteny analyses suggest that they result from teleost whole genome duplication (3R). In contrast to conserved 3R-duplicated ESR2 and GPER, one of 3R-duplicated ESR1 has been lost shortly after teleost emergence. Quantitative PCRs revealed that the five receptors are all widely expressed in the eel, but with differential patterns of tissue expression and regulation. ESR1 only is consistently up-regulated in vivo in female eel BPG-liver axis during induced sexual maturation, and also up-regulated in vitro by estradiol in eel hepatocyte primary cultures. This first comparative study of the five teleost estradiol receptors provides bases for future investigations on differential roles that may have contributed to the conservation of multiple estrogen receptors.
Collapse
Affiliation(s)
- Anne-Gaëlle Lafont
- Muséum National d'Histoire Naturelle, Sorbonne Universités, Research Unit BOREA, Biology of Aquatic Organisms and Ecosystems, CNRS 7208, IRD207, UPMC, UCN, Paris, France.
| | - Karine Rousseau
- Muséum National d'Histoire Naturelle, Sorbonne Universités, Research Unit BOREA, Biology of Aquatic Organisms and Ecosystems, CNRS 7208, IRD207, UPMC, UCN, Paris, France
| | - Jonna Tomkiewicz
- Technical University of Denmark, National Institute of Aquatic Resources, Charlottenlund, Denmark
| | - Sylvie Dufour
- Muséum National d'Histoire Naturelle, Sorbonne Universités, Research Unit BOREA, Biology of Aquatic Organisms and Ecosystems, CNRS 7208, IRD207, UPMC, UCN, Paris, France.
| |
Collapse
|
43
|
Li X, Liu R, Luo L, Yu L, Chen X, Sun L, Wang T, Hylemon PB, Zhou H, Jiang Z, Zhang L. Role of AMP-activated protein kinase α1 in 17α-ethinylestradiol-induced cholestasis in rats. Arch Toxicol 2016; 91:481-494. [PMID: 27090119 DOI: 10.1007/s00204-016-1697-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 03/21/2016] [Indexed: 12/12/2022]
Abstract
Estrogen-induced cholestasis occurs in many women who are susceptible due to pregnancy or hormone replacement therapy for postmenopausal syndrome. 17α-Ethinylestradiol (EE), as a synthetic estrogen, has been widely used to study the underlying mechanisms of estrogen-induced cholestasis. Recent studies have also reported that liver kinase B1 (LKB1)-mediated activation of AMP-activated protein kinase (AMPK) plays a critical role in the regulation of canalicular network formation. However, the role of AMPK in EE-induced cholestasis remains to be determined. In this study, the effects of EE (1-100 µM) on AMPK activation and the expression of farnesoid X receptor (FXR) and hepatic bile acid transporters were examined in in vitro using 3D-cultured rat primary hepatocytes and in in vivo using rat cholestasis models. We also used specific chemical agonist and antagonist of AMPK, AMPK subunit-specific antibodies and lentiviral shRNAs for AMPKα1 and AMPKα2 to delineate the role of AMPK in EE-induced cholestasis and potential cellular mechanisms. We found that EE-induced phosphorylation of AMPKα1 via extracellular signal-regulated kinases-LKB1-mediated signaling pathways and subsequent nuclear translocation accounted for the down-regulation of FXR and bile acid transporters and disruption of bile acid homeostasis. Inhibition of AMPK activation using an AMPK antagonist Compound C (2 µM) or down-regulation of AMPKα1 using gene-specific shRNA attenuated EE-induced cholestasis both in in vitro and in in vivo. In conclusion, these results revealed that activation of cAMP-ERK-LKB1-AMPKα1 signaling pathway plays a critical role in EE-mediated dysregulation of the expression of FXR and bile acid transporters. AMPKα1 may represent an important therapeutic target for estrogen-induced cholestasis.
Collapse
Affiliation(s)
- Xiaojiaoyang Li
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, 210009, Jiangsu, China.,Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, 210009, Jiangsu, China
| | - Runping Liu
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, 210009, Jiangsu, China.,Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Lan Luo
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, 210009, Jiangsu, China
| | - Linxi Yu
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, 210009, Jiangsu, China
| | - Xin Chen
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, 210009, Jiangsu, China
| | - Lixin Sun
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, 210009, Jiangsu, China
| | - Tao Wang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, 210009, Jiangsu, China.,Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, 210009, Jiangsu, China
| | - Phillip B Hylemon
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA, 23298, USA.,McGuire Veterans Affairs Medical Center, Richmond, VA, 23249, USA
| | - Huiping Zhou
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA, 23298, USA. .,McGuire Veterans Affairs Medical Center, Richmond, VA, 23249, USA.
| | - Zhenzhou Jiang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, 210009, Jiangsu, China. .,Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, 210009, Jiangsu, China.
| | - Luyong Zhang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, 210009, Jiangsu, China. .,Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, 210009, Jiangsu, China.
| |
Collapse
|
44
|
Evans NJ, Bayliss AL, Reale V, Evans PD. Characterisation of Signalling by the Endogenous GPER1 (GPR30) Receptor in an Embryonic Mouse Hippocampal Cell Line (mHippoE-18). PLoS One 2016; 11:e0152138. [PMID: 26998610 PMCID: PMC4801207 DOI: 10.1371/journal.pone.0152138] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 03/09/2016] [Indexed: 01/14/2023] Open
Abstract
Estrogen can modulate neuronal development and signalling by both genomic and non-genomic pathways. Many of its rapid, non-genomic effects on nervous tissue have been suggested to be mediated via the activation of the estrogen sensitive G-protein coupled receptor (GPER1 or GPR30). There has been much controversy over the cellular location, signalling properties and endogenous activators of GPER1. Here we describe the pharmacology and signalling properties of GPER1 in an immortalized embryonic hippocampal cell line, mHippoE-18. This cell line does not suffer from the inherent problems associated with the study of this receptor in native tissue or the problems associated with heterologously expression in clonal cell lines. In mHippoE-18 cells, 17β-Estradiol can mediate a dose-dependent rapid potentiation of forskolin-stimulated cyclic AMP levels but does not appear to activate the ERK1/2 pathway. The effect of 17β-Estradiol can be mimicked by the GPER1 agonist, G1, and also by tamoxifen and ICI 182,780 which activate GPER1 in a variety of other preparations. The response is not mimicked by the application of the classical estrogen receptor agonists, PPT, (an ERα agonist) or DPN, (an ERβ agonist), further suggesting that this effect of 17β-Estradiol is mediated through the activation of GPER1. However, after exposure of the cells to the GPER1 specific antagonists, G15 and G36, the stimulatory effects of the above agonists are replaced by dose-dependent inhibitions of forskolin-stimulated cyclic AMP levels. This inhibitory effect is mimicked by aldosterone in a dose-dependent way even in the absence of the GPER1 antagonists. The results are discussed in terms of possible "Biased Antagonism" whereby the antagonists change the conformation of the receptor resulting in changes in the agonist induced coupling of the receptor to different second messenger pathways.
Collapse
Affiliation(s)
- Nicholas J. Evans
- The Signalling Laboratory, The Babraham Institute, Cambridge, CB22 3AT, United Kingdom
| | - Asha L. Bayliss
- The Signalling Laboratory, The Babraham Institute, Cambridge, CB22 3AT, United Kingdom
| | - Vincenzina Reale
- The Signalling Laboratory, The Babraham Institute, Cambridge, CB22 3AT, United Kingdom
| | - Peter D. Evans
- The Signalling Laboratory, The Babraham Institute, Cambridge, CB22 3AT, United Kingdom
| |
Collapse
|
45
|
Gaudet HM, Cheng SB, Christensen EM, Filardo EJ. The G-protein coupled estrogen receptor, GPER: The inside and inside-out story. Mol Cell Endocrinol 2015; 418 Pt 3:207-19. [PMID: 26190834 DOI: 10.1016/j.mce.2015.07.016] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 07/15/2015] [Accepted: 07/15/2015] [Indexed: 02/06/2023]
Abstract
GPER possesses structural and functional characteristics shared by members of the G-protein-coupled receptor (GPCR) superfamily, the largest class of plasma membrane receptors. This newly appreciated estrogen receptor is localized predominately within intracellular membranes in most, but not all, cell types and its surface expression is modulated by steroid hormones and during tissue injury. An intracellular staining pattern is not unique among GPCRs, which employ a diverse array of molecular mechanisms that restrict cell surface expression and effectively regulating receptor binding and activation. The finding that GPER displays an intracellular predisposition has created some confusion as the estrogen-inducible transcription factors, ERα and ERβ, also reside intracellularly, and has led to complex suggestions of receptor interaction. GPER undergoes constitutive retrograde trafficking from the plasma membrane to the endoplasmic reticulum and recent studies indicate its interaction with PDZ binding proteins that sort transmembrane receptors to synaptosomes and endosomes. Genetic targeting and selective ligand approaches as well as cell models that express GPER in the absence of ERs clearly supports GPER as a bonafide "stand alone" receptor. Here, the molecular details that regulate GPER action, its cell biological activities and its implicated roles in physiological and pathological processes are reviewed.
Collapse
Affiliation(s)
- H M Gaudet
- Wheaton College, Department of Chemistry, Norton, MA, 02766, USA
| | - S B Cheng
- Women & Infants Hospital, Brown University, Providence, RI, 02903, USA
| | - E M Christensen
- Wheaton College, Department of Chemistry, Norton, MA, 02766, USA
| | - E J Filardo
- Rhode Island Hospital, Brown University, Providence, RI, 02903, USA.
| |
Collapse
|
46
|
Tokarz J, Möller G, Hrabě de Angelis M, Adamski J. Steroids in teleost fishes: A functional point of view. Steroids 2015; 103:123-44. [PMID: 26102270 DOI: 10.1016/j.steroids.2015.06.011] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 06/11/2015] [Accepted: 06/15/2015] [Indexed: 01/23/2023]
Abstract
Steroid hormones are involved in the regulation of a variety of processes like embryonic development, sex differentiation, metabolism, immune responses, circadian rhythms, stress response, and reproduction in vertebrates. Teleost fishes and humans show a remarkable conservation in many developmental and physiological aspects, including the endocrine system in general and the steroid hormone related processes in particular. This review provides an overview of the current knowledge about steroid hormone biosynthesis and the steroid hormone receptors in teleost fishes and compares the findings to the human system. The impact of the duplicated genome in teleost fishes on steroid hormone biosynthesis and perception is addressed. Additionally, important processes in fish physiology regulated by steroid hormones, which are most dissimilar to humans, are described. We also give a short overview on the influence of anthropogenic endocrine disrupting compounds on steroid hormone signaling and the resulting adverse physiological effects for teleost fishes. By this approach, we show that the steroidogenesis, hormone receptors, and function of the steroid hormones are reasonably well understood when summarizing the available data of all teleost species analyzed to date. However, on the level of a single species or a certain fish-specific aspect of physiology, further research is needed.
Collapse
Affiliation(s)
- Janina Tokarz
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Experimental Genetics, Genome Analysis Center, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany
| | - Gabriele Möller
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Experimental Genetics, Genome Analysis Center, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany
| | - Martin Hrabě de Angelis
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Experimental Genetics, Genome Analysis Center, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany; Lehrstuhl für Experimentelle Genetik, Technische Universität München, 85350 Freising-Weihenstephan, Germany; Member of German Center for Diabetes Research (DZD), Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany
| | - Jerzy Adamski
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Experimental Genetics, Genome Analysis Center, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany; Lehrstuhl für Experimentelle Genetik, Technische Universität München, 85350 Freising-Weihenstephan, Germany; Member of German Center for Diabetes Research (DZD), Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany.
| |
Collapse
|
47
|
Das D, Pal S, Maitra S. Releasing prophase arrest in zebrafish oocyte: synergism between maturational steroid and Igf1. Reproduction 2015; 151:59-72. [PMID: 26500283 DOI: 10.1530/rep-15-0389] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 10/22/2015] [Indexed: 01/09/2023]
Abstract
Binding of 17β-estradiol (E2) to novel G-protein coupled receptor, Gper1, promotes intra-oocyte adenylyl cyclase activity and transactivates epidermal growth factor receptor to ensure prophase-I arrest. Although involvement of either membrane progestin receptor (mPR) or Igf system has been implicated in regulation of meiosis resumption, possibility of concurrent activation and potential synergism between 17α,20β-dihydroxy-4-pregnen-3-one (DHP)- and Igf-mediated signalling cascades in alleviating E2 inhibition of oocyte maturation (OM) has not been investigated. Here using zebrafish (Danio rerio) defolliculated oocytes, we examined the effect of DHP and Igf1, either alone or in combination, in presence or absence of E2, on OM in vitro. While priming of denuded oocytes with E2 blocked spontaneous maturation, co-treatment with DHP (3 nM) and Igf1 (10 nM), but not alone, reversed E2 inhibition and promoted a robust increase in germinal vesicle breakdown (GVBD). Although stimulation with either Igf1 or DHP promoted Akt phosphorylation, pharmacological inhibition of PI3K/Akt signalling prevented Igf1-induced GVBD but delayed DHP action till 4-5 h of incubation. Moreover, high intra-oocyte cAMP attenuates both DHP and Igf1-mediated OM and co-stimulation with DHP and Igf1 could effectively reverse E2 action on PKA phosphorylation. Interestingly, data from in vivo studies reveal that heightened expression of igf1, igf3 transcripts in intact follicles corresponded well with elevated phosphorylation of Igf1r and Akt, mPRa immunoreactivity, PKA inhibition and accelerated GVBD response just prior to ovulation. This indicates potential synergism between maturational steroid and Igf1 which might have physiological relevance in overcoming E2 inhibition of meiosis resumption in zebrafish oocytes.
Collapse
Affiliation(s)
- Debabrata Das
- Department of ZoologyVisva-Bharati University, Santiniketan 731235, India
| | - Soumojit Pal
- Department of ZoologyVisva-Bharati University, Santiniketan 731235, India
| | - Sudipta Maitra
- Department of ZoologyVisva-Bharati University, Santiniketan 731235, India
| |
Collapse
|
48
|
Fitzgerald AC, Peyton C, Dong J, Thomas P. Bisphenol A and Related Alkylphenols Exert Nongenomic Estrogenic Actions Through a G Protein-Coupled Estrogen Receptor 1 (Gper)/Epidermal Growth Factor Receptor (Egfr) Pathway to Inhibit Meiotic Maturation of Zebrafish Oocytes. Biol Reprod 2015; 93:135. [PMID: 26490843 DOI: 10.1095/biolreprod.115.132316] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 10/19/2015] [Indexed: 11/01/2022] Open
Abstract
Xenobiotic estrogens, such as bisphenol A (BPA), disrupt a wide variety of genomic estrogen actions, but their nongenomic estrogen actions remain poorly understood. We investigated nongenomic estrogenic effects of low concentrations of BPA and three related alkylphenols on the inhibition of zebrafish oocye maturation (OM) mediated through a G protein-coupled estrogen receptor 1 (Gper)-dependent epidermal growth factor receptor (Egfr) pathway. BPA (10-100 nM) treatment for 3 h mimicked the effects of estradiol-17beta (E2) and EGF, decreasing spontaneous maturation of defolliculated zebrafish oocytes, an effect not blocked by coincubation with actinomycin D, but blocked by coincubation with a Gper antibody. BPA displayed relatively high binding affinity (15.8% that of E2) for recombinant zebrafish Gper. The inhibitory effects of BPA were attenuated by inhibition of upstream regulators of Egfr, intracellular tyrosine kinase (Src) with PP2, and matrix metalloproteinase with ilomastat. Treatment with an inhibitor of Egfr transactivation, AG1478, and an inhibitor of the mitogen-activated protein kinase (MAPK) 3/1 pathway, U0126, increased spontaneous OM and blocked the inhibitory effects of BPA, E2, and the selective GPER agonist, G-1. Western blot analysis showed that BPA (10-200 nM) mimicked the stimulatory effects of E2 and EGF on Mapk3/1 phosphorylation. Tetrabromobisphenol A, 4-nonylphenol, and tetrachlorobisphenol A (5-100 nM) also inhibited OM, an effect blocked by cotreatment with AG1478, as well as with the GPER antagonist, G-15, and displayed similar binding affinities as BPA to zebrafish Gper. The results suggest that BPA and related alkylphenols disrupt zebrafish OM by a novel nongenomic estrogenic mechanism involving activation of the Gper/Egfr/Mapk3/1 pathway.
Collapse
Affiliation(s)
| | - Candace Peyton
- University of Texas at Austin Marine Science Institute, Port Aransas, Texas
| | - Jing Dong
- University of Texas at Austin Marine Science Institute, Port Aransas, Texas
| | - Peter Thomas
- University of Texas at Austin Marine Science Institute, Port Aransas, Texas
| |
Collapse
|
49
|
Prossnitz ER, Arterburn JB. International Union of Basic and Clinical Pharmacology. XCVII. G Protein-Coupled Estrogen Receptor and Its Pharmacologic Modulators. Pharmacol Rev 2015; 67:505-40. [PMID: 26023144 PMCID: PMC4485017 DOI: 10.1124/pr.114.009712] [Citation(s) in RCA: 193] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Estrogens are critical mediators of multiple and diverse physiologic effects throughout the body in both sexes, including the reproductive, cardiovascular, endocrine, nervous, and immune systems. As such, alterations in estrogen function play important roles in many diseases and pathophysiological conditions (including cancer), exemplified by the lower prevalence of many diseases in premenopausal women. Estrogens mediate their effects through multiple cellular receptors, including the nuclear receptor family (ERα and ERβ) and the G protein-coupled receptor (GPCR) family (GPR30/G protein-coupled estrogen receptor [GPER]). Although both receptor families can initiate rapid cell signaling and transcriptional regulation, the nuclear receptors are traditionally associated with regulating gene expression, whereas GPCRs are recognized as mediating rapid cellular signaling. Estrogen-activated pathways are not only the target of multiple therapeutic agents (e.g., tamoxifen, fulvestrant, raloxifene, and aromatase inhibitors) but are also affected by a plethora of phyto- and xeno-estrogens (e.g., genistein, coumestrol, bisphenol A, dichlorodiphenyltrichloroethane). Because of the existence of multiple estrogen receptors with overlapping ligand specificities, expression patterns, and signaling pathways, the roles of the individual receptors with respect to the diverse array of endogenous and exogenous ligands have been challenging to ascertain. The identification of GPER-selective ligands however has led to a much greater understanding of the roles of this receptor in normal physiology and disease as well as its interactions with the classic estrogen receptors ERα and ERβ and their signaling pathways. In this review, we describe the history and characterization of GPER over the past 15 years focusing on the pharmacology of steroidal and nonsteroidal compounds that have been employed to unravel the biology of this most recently recognized estrogen receptor.
Collapse
Affiliation(s)
- Eric R Prossnitz
- Department of Internal Medicine (E.R.P.) and University of New Mexico Cancer Center (E.R.P., J.B.A.), The University of New Mexico Health Sciences Center, Albuquerque, New Mexico; and Department of Chemistry and Biochemistry, New Mexico State University, Las Cruces, New Mexico (J.B.A.)
| | - Jeffrey B Arterburn
- Department of Internal Medicine (E.R.P.) and University of New Mexico Cancer Center (E.R.P., J.B.A.), The University of New Mexico Health Sciences Center, Albuquerque, New Mexico; and Department of Chemistry and Biochemistry, New Mexico State University, Las Cruces, New Mexico (J.B.A.)
| |
Collapse
|
50
|
Qu T, Zhang SM, Yu LL, Zhang S, Yuan DZ, Xu Q, Zhang JH, He YP, Yue LM. Relocalisation and activation of integrins induced rapidly by oestrogen via G-protein-coupled receptor 30 in mouse blastocysts. Reprod Fertil Dev 2015; 28:RD14227. [PMID: 25950704 DOI: 10.1071/rd14227] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 03/21/2015] [Indexed: 12/20/2022] Open
Abstract
Integrins are the dominant and final adhesion molecules in the attachment process between the blastocysts and endometrium. It is necessary for oestrogen to rapidly activate mouse blastocysts so that they attach to the endometrial epithelium. Our previous study suggested that oestrogen can rapidly induce an increase in intracellular calcium in mouse blastocysts via G-protein-coupled receptor 30 (GPR30). Thus, we deduced that integrins may be involved in GPR30 mediation of the fast effect of oestrogen on mouse blastocysts in implantation. To prove our hypothesis, we used immunofluorescence staining and in vitro coculture of mouse blastocysts and endometrial epithelial cell line (EECs), Ishikawa cells, in the present study. We found that αv and β1 integrin clustered in mouse blastocysts, and that β3 integrin was relocalised to the apical membrane of blastocyst cells when embryos were treated with 1 μM 17β-estradiol (E2), 1 μM E2 conjugated to bovine serum albumin (E2-BSA) and 1 μM G-1, a specific GPR30 agonist, for 30 min respectively, whereas pretreatment with 1 μM G15, a specific GPR30 antagonist, and 5 μM 1,2-Bis(2-aminophenoxy)ethane-N,N,N'',N''-tetraacetic acid tetrakis (acetoxymethyl ester)(BAPTA/AM), a cellular Ca2+ chelator, blocked the localisation of integrins induced by oestrogen via GPR30 in mouse blastocyst cells. E2, E2-BSA and G-1 increased the fibronectin (FN)-binding activity of integrins in blastocysts, whereas G15 and BAPTA/AM blocked the activation of integrins induced by oestrogen via GPR30 in mouse blastocysts. Inhibition of integrins by Arg-Gly-Asp peptide in blastocysts resulted in their failure to adhere to EECs in vitro, even if oestrogen or G-1 was provided. Together, the results indicate the fast effect of oestrogen via the GPR30 membrane receptor further induces relocalisation and activation of integrins in mouse blastocysts, which play important roles in the adhesion of blastocysts to EECs.
Collapse
|