1
|
Peng D, Lu C, Spadacini V, Mitchell K, Tan Y, Zhang D, Levavi-Sivan B, Hu W, Trudeau VL. Hormonal dynamics reveal a stimulatory role for secretoneurin in zebrafish ovulation. PNAS NEXUS 2025; 4:pgaf097. [PMID: 40191135 PMCID: PMC11969067 DOI: 10.1093/pnasnexus/pgaf097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 03/07/2025] [Indexed: 04/09/2025]
Abstract
Surge release of luteinizing hormone (Lh) from the pituitary is essential for fertility as it triggers ovulation. While secretoneurin (SN) is a phylogenetically conserved secretogranin-2-derived peptide that stimulates Lh, its role in ovulation has not been established. To directly compare periovulatory changes in the classical hormones to the emerging reproductive neuropeptides SNa and SNb, simultaneous mass spectrometry measurement of 9 peptides and 5 steroids was conducted in female zebrafish. Regression analysis indicated that levels of SNa1-34 in the brain peaked when type 3 gonadotropin-releasing hormone (Gnrh3) increased (R 2 = 0.71) at the time of the Lh surge, 3.5 h before ovulation. Levels of the naturally occurring derivative SNa1-14 were highest at ovulation, while SNb1-31 was invariable. The bioactivities of SNa1-34 and SNa1-14 were investigated. After injection of SNa1-34 in females that had been isolated from males, 61% (11/18) ovulated within 6 h, which was like the effects of the Lh analog human chorionic gonadotropin (72%; 13/18 females). SNa1-34 injection induces ovulation by increasing time-dependent expression of gnrh3 in the brain, a likely direct stimulation of chorionic gonadotropin alpha (cga) and luteinizing hormone b (lhb) subunit in pituitary, and via the subsequent time-dependent increase in nuclear progesterone receptor (npr) in ovaries. In contrast, SNa1-14 exhibited far fewer effects on gene expression and did not induce ovulation. Our results support the proposal that SN is a reproductive hormone.
Collapse
Affiliation(s)
- Di Peng
- Department of Biology, University of Ottawa, Ottawa K1N 6N5, Ontario, Canada
| | - Chunyu Lu
- Department of Biology, University of Ottawa, Ottawa K1N 6N5, Ontario, Canada
| | - Victoria Spadacini
- Department of Biology, University of Ottawa, Ottawa K1N 6N5, Ontario, Canada
| | - Kimberly Mitchell
- Department of Biology, University of Ottawa, Ottawa K1N 6N5, Ontario, Canada
| | - Yongjun Tan
- Department of Biology, Bioinformatics and Computational Biology Program, College of Arts and Sciences, Saint Louis University, Saint Louis, MO 63103-2010, USA
| | - Dapeng Zhang
- Department of Biology, Bioinformatics and Computational Biology Program, College of Arts and Sciences, Saint Louis University, Saint Louis, MO 63103-2010, USA
| | - Berta Levavi-Sivan
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - Wei Hu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, China
| | - Vance L Trudeau
- Department of Biology, University of Ottawa, Ottawa K1N 6N5, Ontario, Canada
| |
Collapse
|
2
|
Zhou X, Liu W, Cong B, Deng A, Lin J, Zhao L, Liu S. Transcriptomics-based analysis of neurotoxic and reproductive effects in turbot (Scophthalmus maximus) after exposure to tris (2-chloroethyl) phosphate (TCEP). BMC Genomics 2025; 26:38. [PMID: 39815191 PMCID: PMC11734547 DOI: 10.1186/s12864-024-11061-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 11/18/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Tris (2-chloroethyl) phosphate (TCEP), a widely used flame retardant, is widespread in the environment and potentially harmful to organisms. However, the specific mechanisms of TCEP-induced neurological and reproductive toxicity in fish are largely unknown. Turbot (Scophthalmus maximus) is cultivated on a large scale, and the emergence of pollutants with endocrine disrupting effects seriously affects its economic benefits. This study aimed to investigate the toxic effects of TCEP on turbot by integrating physio-biochemical and transcriptomic analyses. RESULTS TCEP exposure induced severe neuroendocrine disrupting effects in turbot. Firstly, the hormone levels of luteinizing hormone (LH), follicle-stimulating hormone (FSH), estradiol (E2), and 11-ketotestosterone (11-KT) were significantly decreased under prolonged TCEP stress, which may have a negative impact on normal reproductive function. We identified and summarized representative differentially expressed genes (DEGs) and their functions, such as endocrine system and oxidative stress. Pathway enrichment showed that the toxicological characteristics of TCEP on turbot were neuroendocrine regulation disorders, including oxidative phosphorylation, apoptosis, steroid biosynthesis, GnRH signaling pathway and so on. Weighted gene co-expression network analysis (WGCNA) also revealed key genes involved in these pathways. Among these genes, those encoding the components of the electron transport chain presented an initial increase in expression followed by a decrease, indicating that TCEP stress might affect mitochondrial function and lead to cell damage. This finding was also supported by the upregulation of apoptosis-related gene expression. Moreover, acute exposure to TCEP regulated MAPK-mediated transduction and regulation of GnRH signaling, thereby altering the expression of hypothalamic-pituitary-gonadal (HPG) axis-related genes. CONCLUSIONS These findings revealed the endocrine disrupting effects of TCEP on turbot and identified biomarkers related to reproductive toxicity, providing early warning for the monitoring of healthy aquaculture.
Collapse
Affiliation(s)
- Xiaoqian Zhou
- School of Advanced Manufacturing School of Ocean, Fuzhou University, Jinjiang, 362200, China
| | - Weifeng Liu
- Laboratory for Marine Ecology and Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, 266061, China
| | - Bailin Cong
- School of Advanced Manufacturing School of Ocean, Fuzhou University, Jinjiang, 362200, China.
- Laboratory for Marine Ecology and Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, 266061, China.
| | - Aifang Deng
- Laboratory for Marine Ecology and Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, 266061, China
| | - Jing Lin
- Laboratory for Marine Ecology and Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, 266061, China
| | - Linlin Zhao
- Laboratory for Marine Ecology and Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, 266061, China
| | - Shenghao Liu
- Laboratory for Marine Ecology and Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, 266061, China
| |
Collapse
|
3
|
Hollander-Cohen L, Cohen O, Shulman M, Aiznkot T, Fontanaud P, Revah O, Mollard P, Golan M, Levavi-Sivan B. The satiety hormone cholecystokinin gates reproduction in fish by controlling gonadotropin secretion. eLife 2024; 13:RP96344. [PMID: 39717899 DOI: 10.7554/elife.96344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2024] Open
Abstract
Life histories of oviparous species dictate high metabolic investment in the process of gonadal development leading to ovulation. In vertebrates, these two distinct processes are controlled by the gonadotropins follicle-stimulating hormone (FSH) and luteinizing hormone (LH), respectively. While it was suggested that a common secretagogue, gonadotropin-releasing hormone (GnRH), oversees both functions, the generation of loss-of-function fish challenged this view. Here, we reveal that the satiety hormone cholecystokinin (CCK) is the primary regulator of this axis in zebrafish. We found that FSH cells express a CCK receptor, and our findings demonstrate that mutating this receptor results in a severe hindrance to ovarian development. Additionally, it causes a complete shutdown of both gonadotropins secretion. Using in-vivo and ex-vivo calcium imaging of gonadotrophs, we show that GnRH predominantly activates LH cells, whereas FSH cells respond to CCK stimulation, designating CCK as the bona fide FSH secretagogue. These findings indicate that the control of gametogenesis in fish was placed under different neural circuits, that are gated by CCK.
Collapse
Affiliation(s)
- Lian Hollander-Cohen
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, Hebrew University of Jerusalem, Rehovot, Israel
| | - Omer Cohen
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, Hebrew University of Jerusalem, Rehovot, Israel
| | - Miriam Shulman
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, Hebrew University of Jerusalem, Rehovot, Israel
| | - Tomer Aiznkot
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, Hebrew University of Jerusalem, Rehovot, Israel
| | - Pierre Fontanaud
- Institute of Functional Genomics, University of Montpellier, Montpellier, France
- BioCampus Montpellier, University of Montpellier, Montpellier, France
| | - Omer Revah
- The Koret School of Veterinary Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Patrice Mollard
- Institute of Functional Genomics, University of Montpellier, Montpellier, France
- BioCampus Montpellier, University of Montpellier, Montpellier, France
| | - Matan Golan
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, Hebrew University of Jerusalem, Rehovot, Israel
- Department of Poultry and Aquaculture, Institute of Animal Sciences, Agricultural Research Organization, Volcani Center, Rishon Letziyon, Israel
| | - Berta Levavi-Sivan
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, Hebrew University of Jerusalem, Rehovot, Israel
| |
Collapse
|
4
|
Royan MR, Hodne K, Nourizadeh-Lillabadi R, Weltzien FA, Henkel C, Fontaine R. Day length regulates gonadotrope proliferation and reproduction via an intra-pituitary pathway in the model vertebrate Oryzias latipes. Commun Biol 2024; 7:388. [PMID: 38553567 PMCID: PMC10980775 DOI: 10.1038/s42003-024-06059-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 03/16/2024] [Indexed: 04/01/2024] Open
Abstract
In seasonally breeding mammals and birds, the production of the hormones that regulate reproduction (gonadotropins) is controlled by a complex pituitary-brain-pituitary pathway. Indeed, the pituitary thyroid-stimulating hormone (TSH) regulates gonadotropin expression in pituitary gonadotropes, via dio2-expressing tanycytes, hypothalamic Kisspeptin, RFamide-related peptide, and gonadotropin-releasing hormone neurons. However, in fish, how seasonal environmental signals influence gonadotropins remains unclear. In addition, the seasonal regulation of gonadotrope (gonadotropin-producing cell) proliferation in the pituitary is, to the best of our knowledge, not elucidated in any vertebrate group. Here, we show that in the vertebrate model Japanese medaka (Oryzias latipes), a long day seasonally breeding fish, photoperiod (daylength) not only regulates hormone production by the gonadotropes but also their proliferation. We also reveal an intra-pituitary pathway that regulates gonadotrope cell number and hormone production. In this pathway, Tsh regulates gonadotropes via folliculostellate cells within the pituitary. This study suggests the existence of an alternative regulatory mechanism of seasonal gonadotropin production in fish.
Collapse
Affiliation(s)
- Muhammad Rahmad Royan
- Department of Preclinical Science and Pathology, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| | - Kjetil Hodne
- Department of Preclinical Science and Pathology, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| | - Rasoul Nourizadeh-Lillabadi
- Department of Preclinical Science and Pathology, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| | - Finn-Arne Weltzien
- Department of Preclinical Science and Pathology, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| | - Christiaan Henkel
- Department of Preclinical Science and Pathology, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| | - Romain Fontaine
- Department of Preclinical Science and Pathology, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway.
| |
Collapse
|
5
|
Kazeto Y, Ito R, Tanaka T, Suzuki H, Ozaki Y, Okuzawa K, Gen K. Establishment of cell-lines stably expressing recombinant Japanese eel follicle-stimulating hormone and luteinizing hormone using CHO-DG44 cells: fully induced ovarian development at different modes. Front Endocrinol (Lausanne) 2023; 14:1201250. [PMID: 37693354 PMCID: PMC10486264 DOI: 10.3389/fendo.2023.1201250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/24/2023] [Indexed: 09/12/2023] Open
Abstract
The gonadotropins (Gth), follicle-stimulating hormone (Fsh) and luteinizing hormone (Lh), play central roles in gametogenesis in vertebrates. However, available information on their differential actions in teleost, especially in vivo, is insufficient. In this study, we established stable CHO-DG44 cell lines expressing long-lasting recombinant Japanese eel Fsh and Lh with extra O-glycosylation sites (Fsh-hCTP and Lh-hCTP), which were produced in abundance. Immature female eels received weekly intraperitoneal injections of Gths. Fsh-hCTP induced the entire ovarian development by 8 weeks from the beginning of injection; thus, the ovaries of most fish were at the migratory nucleus stage while the same stage was observed in eels after 4 weeks in the Lh-hCTP-treated group. In contrast, all pretreated and saline-injected eels were in the pre-vitellogenic stage. Gonadosomatic indices in the Fsh-hCTP-treated group were significantly higher than those in the Lh-hCTP group at the migratory nucleus stage because of the significantly higher frequency of advanced ovarian follicles. Ovarian mRNA levels of genes related to E2 production (cyp11a1, cyp17a1, cyp19a1, hsd3b, fshr, and lhr) were measured using real-time quantitative reverse transcription-polymerase chain reaction (RT-PCR). All genes were induced by both Fsh-hCTP and Lh-hCTP, with a peak at either the mid- or late vitellogenic stages. Transcript abundance of cyp19a1 and fshr in the Lh-hCTP group were significantly higher than those in the Fsh-hCTP group, whereas no difference in the expression of other genes was observed between the groups. Fluctuations in serum levels of sex steroid hormones (estradiol-17β, 11-ketotestosterone, and testosterone) in female eels were comparable in the Fsh-hCTP and Lh-hCTP groups, thus increasing toward the maturational phase. Furthermore, the fecundity of the eels induced to mature by Fsh-hCTP was significantly higher than that induced by Lh-hCTP. These findings indicate that Fsh and Lh can induce ovarian development in distinctively different modes in the Japanese eel.
Collapse
Affiliation(s)
- Yukinori Kazeto
- Fisheries Technology Institute, Minamiizu Field Station, Japan Fisheries Research and Education Agency, Minamiizu, Shizuoka, Japan
| | - Risa Ito
- Fisheries Technology Institute, Tamaki Field Station, Japan Fisheries Research and Education Agency, Tamaki, Mie, Japan
| | - Toshiomi Tanaka
- Hamanako Branch, Shizuoka Prefectural Research Institute of Fishery and Ocean, Hamamatsu, Shizuoka, Japan
| | - Hiroshi Suzuki
- Fisheries Technology Institute, Shibushi Field Station, Japan Fisheries Research and Education Agency, Shibushi, Kagoshima, Japan
| | - Yuichi Ozaki
- Fisheries Technology Institute, Tamaki Field Station, Japan Fisheries Research and Education Agency, Tamaki, Mie, Japan
| | - Koichi Okuzawa
- Fisheries Technology Institute, Tamaki Field Station, Japan Fisheries Research and Education Agency, Tamaki, Mie, Japan
| | - Koichiro Gen
- Fisheries Technology Institute, Nagasaki Station, Japan Fisheries Research and Education Agency, Nagasaki, Japan
| |
Collapse
|
6
|
Kho KH, Sukhan ZP, Yang SW, Hwang NY, Lee WK. Gonadotropins and Sex Steroid Hormones in Captive-Reared Small Yellow Croaker ( Larimichthys polyactis) and Their Role in Female Reproductive Dysfunction. Int J Mol Sci 2023; 24:ijms24108919. [PMID: 37240265 DOI: 10.3390/ijms24108919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/10/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
The seed production of small yellow croaker (SYC) is constrained by reproductive dysfunction in captive-reared females. Reproductive dysfunction is closely linked to endocrine reproductive mechanisms. To better understand the reproductive dysfunction in captive broodstock, functional characterization of gonadotropins (GtHs: follicle stimulating hormone β subunit, fshβ; luteinizing hormone β subunit, lhβ; and glycoprotein α subunit, gpα) and sex steroids (17β-estradiol, E2; testosterone, T; progesterone; P) was performed using qRT-PCR, ELISA, in vivo, and in-vitro assay. The pituitary GtHs and gonadal steroids levels were significantly higher in ripen fish of both sexes. However, changes in lhβ and E2 levels in females were not significant in the developing and ripen stages. Furthermore, GtHs and steroids levels were lower in females compared to males throughout the reproductive cycle. In vivo administration of gonadotropin releasing hormone analogue (GnRHa) significantly increased the expression of GtHs in both dose- and time-related manners. The lower and higher doses of GnRHa led to successful spawning in male and female SYC, respectively. Sex steroids in vitro significantly inhibited the expression of lhβ in female SYC. Overall, GtHs were shown to play a vital role in final gonadal maturation, while steroids promoted negative feedback in the regulation of pituitary GtHs. Lower levels of GtHs and steroids might be key components in the reproductive dysfunction of captive-reared female SYC.
Collapse
Affiliation(s)
- Kang Hee Kho
- Department of Fisheries Science, Chonnam National University, Yeosu 59626, Republic of Korea
| | - Zahid Parvez Sukhan
- Department of Fisheries Science, Chonnam National University, Yeosu 59626, Republic of Korea
| | - Seok-Woo Yang
- Ocean and Fisheries Science Institute, Jeollanam-do 59326, Republic of Korea
| | - Nam-Yong Hwang
- Ocean and Fisheries Science Institute, Jeollanam-do 59326, Republic of Korea
| | - Won-Kyo Lee
- Department of Fisheries Science, Chonnam National University, Yeosu 59626, Republic of Korea
| |
Collapse
|
7
|
Chen W, Zhai Y, Zhu B, Wu K, Fan Y, Zhou X, Liu L, Ge W. Loss of growth differentiation factor 9 causes an arrest of early folliculogenesis in zebrafish-A novel insight into its action mechanism. PLoS Genet 2022; 18:e1010318. [PMID: 36520929 PMCID: PMC9799306 DOI: 10.1371/journal.pgen.1010318] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 12/29/2022] [Accepted: 12/02/2022] [Indexed: 12/23/2022] Open
Abstract
Growth differentiation factor 9 (GDF9) was the first oocyte-specific growth factor identified; however, most information about GDF9 functions comes from studies in the mouse model. In this study, we created a mutant for Gdf9 gene (gdf9-/-) in zebrafish using TALEN approach. The loss of Gdf9 caused a complete arrest of follicle development at primary growth (PG) stage. These follicles eventually degenerated, and all mutant females gradually changed to males through sex reversal, which could be prevented by mutation of the male-promoting gene dmrt1. Interestingly, the phenotypes of gdf9-/- could be rescued by simultaneous mutation of inhibin α (inha-/-) but not estradiol treatment, suggesting a potential role for the activin-inhibin system or its signaling pathway in Gdf9 actions. In gdf9-null follicles, the expression of activin βAa (inhbaa), but not βAb (inhbab) and βB (inhbb), decreased dramatically; however, its expression rebounded in the double mutant (gdf9-/-;inha-/-). These results indicate clearly that the activation of PG follicles to enter the secondary growth (SG) requires intrinsic factors from the oocyte, such as Gdf9, which in turn works on the neighboring follicle cells to trigger follicle activation, probably involving activins. In addition, our data also support the view that estrogens are not involved in follicle activation as recently reported.
Collapse
Affiliation(s)
- Weiting Chen
- Department of Biomedical Sciences and Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Yue Zhai
- Department of Biomedical Sciences and Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Bo Zhu
- Department of Biomedical Sciences and Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Kun Wu
- Department of Biomedical Sciences and Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Yuqin Fan
- Department of Biomedical Sciences and Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Xianqing Zhou
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, Beijing, China
| | - Lin Liu
- School of Life Science, South China Normal University, Guangzhou, China
| | - Wei Ge
- Department of Biomedical Sciences and Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| |
Collapse
|
8
|
Chen J, Katada Y, Okimura K, Yamaguchi T, Guh YJ, Nakayama T, Maruyama M, Furukawa Y, Nakane Y, Yamamoto N, Sato Y, Ando H, Sugimura A, Tabata K, Sato A, Yoshimura T. Prostaglandin E 2 synchronizes lunar-regulated beach spawning in grass puffers. Curr Biol 2022; 32:4881-4889.e5. [PMID: 36306789 DOI: 10.1016/j.cub.2022.09.062] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 08/11/2022] [Accepted: 09/29/2022] [Indexed: 11/22/2022]
Abstract
Many organisms living along the coastlines synchronize their reproduction with the lunar cycle. At the time of spring tide, thousands of grass puffers (Takifugu alboplumbeus) aggregate and vigorously tremble their bodies at the water's edge to spawn. To understand the mechanisms underlying this spectacular semilunar beach spawning, we collected the hypothalamus and pituitary from male grass puffers every week for 2 months. RNA sequencing (RNA-seq) analysis identified 125 semilunar genes, including genes crucial for reproduction (e.g., gonadotropin-releasing hormone 1 [gnrh1], luteinizing hormone β subunit [lhb]) and receptors for pheromone prostaglandin E (PGE). PGE2 is secreted into the seawater during the spawning, and its administration activates olfactory sensory neurons and triggers trembling behavior of surrounding individuals. These results suggest that PGE2 synchronizes lunar-regulated beach-spawning behavior in grass puffers. To further explore the mechanism that regulates the lunar-synchronized transcription of semilunar genes, we searched for semilunar transcription factors. Spatial transcriptomics and multiplex fluorescent in situ hybridization showed co-localization of the semilunar transcription factor CCAAT/enhancer-binding protein δ (cebpd) and gnrh1, and cebpd induced the promoter activity of gnrh1. Taken together, our study demonstrates semilunar genes that mediate lunar-synchronized beach-spawning behavior. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Junfeng Chen
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Aichi, Japan; Laboratory of Animal Integrative Physiology, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Aichi, Japan
| | - Yuma Katada
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Aichi, Japan; Laboratory of Animal Integrative Physiology, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Aichi, Japan
| | - Kousuke Okimura
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Aichi, Japan; Laboratory of Animal Integrative Physiology, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Aichi, Japan
| | - Taiki Yamaguchi
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Aichi, Japan; Laboratory of Animal Integrative Physiology, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Aichi, Japan
| | - Ying-Jey Guh
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Aichi, Japan; Laboratory of Animal Integrative Physiology, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Aichi, Japan
| | - Tomoya Nakayama
- Laboratory of Animal Integrative Physiology, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Aichi, Japan; Institute for Advanced Research, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Aichi, Japan
| | - Michiyo Maruyama
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Aichi, Japan; Laboratory of Animal Integrative Physiology, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Aichi, Japan
| | - Yuko Furukawa
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Aichi, Japan; Laboratory of Animal Integrative Physiology, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Aichi, Japan
| | - Yusuke Nakane
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Aichi, Japan; Laboratory of Animal Integrative Physiology, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Aichi, Japan
| | - Naoyuki Yamamoto
- Laboratory of Fish Biology, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Aichi, Japan
| | - Yoshikatsu Sato
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Aichi, Japan
| | - Hironori Ando
- Sado Marine Biological Station, Sado Island Center for Ecological Sustainability, Niigata University, 87 Tassha, Sado 952-2135, Niigata, Japan
| | - Asako Sugimura
- Toyota Boshoku Corporation, 1-1 Toyoda-cho, Kariya 448-8651, Aichi, Japan
| | - Kazufumi Tabata
- Toyota Boshoku Corporation, 1-1 Toyoda-cho, Kariya 448-8651, Aichi, Japan
| | - Ayato Sato
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Aichi, Japan
| | - Takashi Yoshimura
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Aichi, Japan; Laboratory of Animal Integrative Physiology, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Aichi, Japan.
| |
Collapse
|
9
|
Ramos-Júdez S, Danis T, Angelova N, Tsakogiannis A, Giménez I, Tsigenopoulos CS, Duncan N, Manousaki T. Transcriptome analysis of flathead grey mullet ( Mugil cephalus) ovarian development induced by recombinant gonadotropin hormones. Front Physiol 2022; 13:1033445. [PMID: 36388126 PMCID: PMC9664002 DOI: 10.3389/fphys.2022.1033445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/14/2022] [Indexed: 06/16/2023] Open
Abstract
Background: Treatment with recombinant gonadotropin hormones (rGths), follicle-stimulating hormone (rFsh) and luteinizing hormone (rLh), was shown to induce and complete vitellogenesis to finally obtain viable eggs and larvae in the flathead grey mullet (Mugil cephalus), a teleost arrested at early stages of gametogenesis in intensive captivity conditions. This study aimed to investigate the transcriptomic changes that occur in the ovary of females during the rGths-induced vitellogenesis. Methods: Ovarian samples were collected through biopsies from the same five females at four stages of ovarian development. RNASeq libraries were constructed for all stages studied, sequenced on an Illumina HiSeq4000, and a de novo transcriptome was constructed. Differentially expressed genes (DEGs) were identified between stages and the functional properties of DEGs were characterized by comparison with the gene ontology and Kyoto Encyclopedia. An enrichment analysis of molecular pathways was performed. Results: The de novo transcriptome comprised 287,089 transcripts after filtering. As vitellogenesis progressed, more genes were significantly upregulated than downregulated. The rFsh application induced ovarian development from previtellogenesis to early-to-mid-vitellogenesis with associated pathways enriched from upregulated DEGs related to ovarian steroidogenesis and reproductive development, cholesterol metabolism, ovarian growth and differentiation, lipid accumulation, and cell-to-cell adhesion pathways. The application of rFsh and rLh at early-to-mid-vitellogenesis induced the growth of oocytes to late-vitellogenesis and, with it, the enrichment of pathways from upregulated DEGs related to the production of energy, such as the lysosomes activity. The application of rLh at late-vitellogenesis induced the completion of vitellogenesis with the enrichment of pathways linked with the switch from vitellogenesis to oocyte maturation. Conclusion: The DEGs and enriched molecular pathways described during the induced vitellogenesis of flathead grey mullet with rGths were typical of natural oogenesis reported for other fish species. Present results add new knowledge to the rGths action to further raise the possibility of using rGths in species that present similar reproductive disorders in aquaculture, the aquarium industry as well as the conservation of endangered species.
Collapse
Affiliation(s)
| | - Theodoros Danis
- Institute of Marine Biology, Biotechnology and Aquaculture (IMBBC), Hellenic Centre for Marine Research (H.C.M.R.), Heraklion, Greece
| | - Nelina Angelova
- Institute of Marine Biology, Biotechnology and Aquaculture (IMBBC), Hellenic Centre for Marine Research (H.C.M.R.), Heraklion, Greece
| | - Alexandros Tsakogiannis
- Institute of Marine Biology, Biotechnology and Aquaculture (IMBBC), Hellenic Centre for Marine Research (H.C.M.R.), Heraklion, Greece
| | | | - Costas S. Tsigenopoulos
- Institute of Marine Biology, Biotechnology and Aquaculture (IMBBC), Hellenic Centre for Marine Research (H.C.M.R.), Heraklion, Greece
| | | | - Tereza Manousaki
- Institute of Marine Biology, Biotechnology and Aquaculture (IMBBC), Hellenic Centre for Marine Research (H.C.M.R.), Heraklion, Greece
| |
Collapse
|
10
|
The N-Linked Glycosylation Site N191 Is Necessary for PKA Signal Transduction in Eel Follicle-Stimulating Hormone Receptor. Int J Mol Sci 2022; 23:ijms232112792. [PMID: 36361582 PMCID: PMC9655291 DOI: 10.3390/ijms232112792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 11/24/2022] Open
Abstract
The follicle-stimulating hormone receptor (FSHR) contains several N-linked glycosylation sites in its extracellular region. We conducted the present study to determine whether conserved glycosylated sites in eel FSHR are necessary for cyclic adenosine monophosphate (cAMP) signal transduction. We used site-directed mutagenesis to induce four mutations (N120Q, N191Q, N272Q, and N288Q) in the N-linked glycosylation sites of eel FSHR. In the eel FSHR wild-type (wt), the cAMP response was gradually increased in a dose-dependent manner (0.01–1500 ng/mL), displaying a high response (approximately 57.5 nM/104 cells) at the Rmax level. Three mutants (N120Q, N272Q, and N288Q) showed a considerably decreased signal transduction as a result of high-ligand treatment, whereas one mutant (N191Q) exhibited a completely impaired signal transduction. The expression level of the N191Q mutant was only 9.2% relative to that of the eel FSHR-wt, indicating a negligible expression level. The expression levels of the N120Q and N272Q mutants were approximately 35.9% and 24% of the FSHG-wt, respectively. The N288Q mutant had an expression level similar to that of the eel FSHR-wt, despite the mostly impaired cAMP responsiveness. The loss of the cell surface agonist-receptor complexes was very rapid in the cells expressing eel FSHR-wt and FSHR-N288Q mutants. Specifically, the N191Q mutant was completely impaired by the loss of cell surface receptors, despite treatment with a high concentration of the agonist. Therefore, we suggest that the N191 site is necessary for cAMP signal transduction. This finding implies that the cAMP response, mediated by G proteins, is directly related to the loss of cell surface receptors as a result of high-agonist treatment.
Collapse
|
11
|
The Zebrafish, an Outstanding Model for Biomedical Research in the Field of Melatonin and Human Diseases. Int J Mol Sci 2022; 23:ijms23137438. [PMID: 35806441 PMCID: PMC9267299 DOI: 10.3390/ijms23137438] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/30/2022] [Accepted: 07/01/2022] [Indexed: 02/06/2023] Open
Abstract
The zebrafish has become an excellent model for the study of human diseases because it offers many advantages over other vertebrate animal models. The pineal gland, as well as the biological clock and circadian rhythms, are highly conserved in zebrafish, and melatonin is produced in the pineal gland and in most organs and tissues of the body. Zebrafish have several copies of the clock genes and of aanat and asmt genes, the latter involved in melatonin synthesis. As in mammals, melatonin can act through its membrane receptors, as with zebrafish, and through mechanisms that are independent of receptors. Pineal melatonin regulates peripheral clocks and the circadian rhythms of the body, such as the sleep/wake rhythm, among others. Extrapineal melatonin functions include antioxidant activity, inducing the endogenous antioxidants enzymes, scavenging activity, removing free radicals, anti-inflammatory activity through the regulation of the NF-κB/NLRP3 inflammasome pathway, and a homeostatic role in mitochondria. In this review, we introduce the utility of zebrafish to analyze the mechanisms of action of melatonin. The data here presented showed that the zebrafish is a useful model to study human diseases and that melatonin exerts beneficial effects on many pathophysiological processes involved in these diseases.
Collapse
|
12
|
Qin M, Xie Q, Wu K, Zhou X, Ge W. Loss of Nobox prevents ovarian differentiation from juvenile ovaries in zebrafish. Biol Reprod 2022; 106:1254-1266. [PMID: 35157068 DOI: 10.1093/biolre/ioac036] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/10/2022] [Accepted: 02/12/2022] [Indexed: 11/12/2022] Open
Abstract
As a species without master sex-determining genes, zebrafish displays high plasticity in sex differentiation, making it an excellent model for studying the regulatory mechanisms underlying gonadal differentiation and gametogenesis. Despite being a gonochorist, zebrafish is a juvenile hermaphrodite that undergoes a special phase of juvenile ovary before further differentiation into functional testis and ovary. The mechanisms underlying juvenile ovary formation and subsequent gonadal differentiation remain largely unknown. In a recent study, we demonstrated an important role for Figla (factor in the germline alpha) in zebrafish oogenesis. In this study, we explored the role of Nobox/nobox (new born ovary homeobox protein), another oocyte-specific transcription factor in females, in early zebrafish gonadogenesis using CRISPR/Cas9 technology. As in mammals, nobox is specifically expressed in zebrafish gonads with a dimorphic pattern at juvenile stage. In contrast to the mutant of figla (another oocyte-specific transcription factor), the nobox mutants showed formation of typical perinucleolar (PN) follicles at primary growth (PG) stage in juvenile gonads, suggesting occurrence of follicle assembly from cystic oocytes (chromatin nucleolar stage, CN). These follicles, however, failed to develop further to form functional ovaries, resulting in all-male phenotype. Despite its expression in adult testis, the loss of nobox did not seem to affect testis development, spermatogenesis and male spawning. In summary, our results indicate an important role for Nobox in zebrafish ovarian differentiation and early folliculogenesis.
Collapse
Affiliation(s)
- Mingming Qin
- Department of Biomedical Sciences and Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Qingping Xie
- Department of Biomedical Sciences and Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau, China.,Institute of Hydrobiology, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Kun Wu
- Department of Biomedical Sciences and Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau, China.,State Key Laboratory for Biocontrol, Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, School of Marine Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xianqing Zhou
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, Beijing, China
| | - Wei Ge
- Department of Biomedical Sciences and Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| |
Collapse
|
13
|
Roles of Gonadotropin Receptors in Sexual Development of Medaka. Cells 2022; 11:cells11030387. [PMID: 35159197 PMCID: PMC8834109 DOI: 10.3390/cells11030387] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/17/2022] [Accepted: 01/20/2022] [Indexed: 12/10/2022] Open
Abstract
The gonadotropins, follicle-stimulating hormone (FSH) and luteinizing hormone (LH), are secreted from the pituitary and bind to the FSH receptor (FSHR) and LH receptor (LHR) to regulate gonadal development in vertebrates. Previously, using fshr-knockout (KO) medaka (Oryzias latipes), we demonstrated that FSH regulates ovarian development by elevating estrogen levels. However, the lhr-KO phenotype in medaka is poorly characterized. Here, we generated lhr-KO medaka using the transcription activator-like effector nuclease (TALEN) technique. We analyzed its phenotype and that of fshr-KO, lhr;fshr double-heterozygotes (double-hetero), and double-KO fish. All genetically male medaka displayed normal testes and were fertile, whereas fshr-KO and double-KO genetically female fish displayed small ovaries containing many early pre-vitellogenic oocytes and were infertile. Although lhr-KO genetically female fish had normal ovaries with full-grown oocytes, ovulation did not occur. Levels of 17α,20β-dihydroxy-4-pregnen-3-one, which is required for meiotic maturation of oocytes and sperm maturation in teleost fish, were significantly decreased in all KO female medaka ovaries except for double-heteros. Further, 17β-estradiol levels in fshr-KO and double-KO ovaries were significantly lower than those in double-heteros. These findings indicate that LH is necessary for oocyte maturation and FSH is necessary for follicle development, but that neither are essential for spermatogenesis in medaka.
Collapse
|
14
|
Molés G, Hausken K, Carrillo M, Zanuy S, Levavi-Sivan B, Gómez A. Generation and use of recombinant gonadotropins in fish. Gen Comp Endocrinol 2020; 299:113555. [PMID: 32687933 DOI: 10.1016/j.ygcen.2020.113555] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 07/11/2020] [Accepted: 07/14/2020] [Indexed: 02/09/2023]
Abstract
Understanding the differential roles of the pituitary gonadotropins Fsh and Lh in gonad maturation is crucial for a successful manipulation of the reproductive process in fish, and requires species-specific tools and appropriate active hormones. With the increasing availability of fish cDNAs coding for gonadotropin subunits, the production of recombinant hormones in heterologous systems has gradually substituted the approach of isolating native hormones. These recombinant hormones can be continually produced without depending on the fish as starting material and no cross-contamination with other pituitary glycoproteins is assured. Recombinant gonadotropins should be produced in eukaryotic cells, which have glycosylation capacity, but this post-translational modification varies greatly depending on the cell system, influencing hormone activity and stability. The production of recombinant gonadotropin beta-subunits to be used as antigens for antibody production has allowed the development of immunoassays for quantification of gonadotropins in some fish species. The administration in vivo of dimeric homologous recombinant gonadotropins has been used in basic studies and as a biotechnological approach to induce gametogenesis. In addition, gene-based therapies using somatic transfer of the gonadotropin genes have been tested as an alternative for hormone delivery in vivo. In summary, the use of homologous hormonal treatments can open new strategies in aquaculture to solve reproductive problems or develop out-of-season breeding programs.
Collapse
Affiliation(s)
- G Molés
- Instituto de Acuicultura Torre de la Sal, Consejo Superior de Investigaciones Científicas (CSIC), Ribera de Cabanes s/n, 12595 Castelló, Spain
| | - K Hausken
- The Robert H. Smith Faculty of Agriculture, Food and Environment, Department of Animal Sciences, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - M Carrillo
- Instituto de Acuicultura Torre de la Sal, Consejo Superior de Investigaciones Científicas (CSIC), Ribera de Cabanes s/n, 12595 Castelló, Spain
| | - S Zanuy
- Instituto de Acuicultura Torre de la Sal, Consejo Superior de Investigaciones Científicas (CSIC), Ribera de Cabanes s/n, 12595 Castelló, Spain
| | - B Levavi-Sivan
- The Robert H. Smith Faculty of Agriculture, Food and Environment, Department of Animal Sciences, The Hebrew University of Jerusalem, Rehovot 76100, Israel.
| | - A Gómez
- Instituto de Acuicultura Torre de la Sal, Consejo Superior de Investigaciones Científicas (CSIC), Ribera de Cabanes s/n, 12595 Castelló, Spain.
| |
Collapse
|
15
|
Zhang Z, Wu K, Ren Z, Ge W. Genetic evidence for Amh modulation of gonadotropin actions to control gonadal homeostasis and gametogenesis in zebrafish and its noncanonical signaling through Bmpr2a receptor. Development 2020; 147:dev189811. [PMID: 33060133 DOI: 10.1242/dev.189811] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 10/12/2020] [Indexed: 12/11/2022]
Abstract
Anti-Müllerian hormone (Amh) plays an important role in gonadal function. Amh deficiency causes severe gonadal dysgenesis and dysfunction in zebrafish, with gonadal hypertrophy in both sexes. However, its mechanism of action remains unknown. Intriguingly, the Amh cognate type II receptor (Amhr2) is missing in the zebrafish genome, in sharp contrast to other species. Using a series of zebrafish mutants (amh, fshb, fshr and lhcgr), we provided unequivocal evidence for actions of Amh, via modulation of gonadotropin signaling, on both germ cell proliferation and differentiation. The gonadal hypertrophy in amh mutants was abolished in the absence of Fshr in females or Fshr/Lhcgr in males. Furthermore, we demonstrated that knockout of bmpr2a, but not bmpr2b, phenocopied all phenotypes of the amh mutant in both sexes, including gonadal hypertrophy, hyperproliferation of germ cells, retarded gametogenesis and reduced fshb expression. In summary, the present study provided comprehensive genetic evidence for an intimate interaction of gonadotropin and Amh pathways in gonadal homeostasis and gametogenesis and for Bmpr2a as the possible missing link for Amh signaling in zebrafish.
Collapse
Affiliation(s)
- Zhiwei Zhang
- Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Kun Wu
- Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Zhiqin Ren
- Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Wei Ge
- Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| |
Collapse
|
16
|
Zhang Z, Zhu B, Chen W, Ge W. Anti-Müllerian hormone (Amh/amh) plays dual roles in maintaining gonadal homeostasis and gametogenesis in zebrafish. Mol Cell Endocrinol 2020; 517:110963. [PMID: 32745576 DOI: 10.1016/j.mce.2020.110963] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 11/21/2022]
Abstract
Anti-Müllerian hormone (AMH/Amh) plays a role in gonadal differentiation and function across vertebrates. In zebrafish we demonstrated that Amh deficiency caused severe gonadal dysgenesis and dysfunction. The mutant gonads showed extreme hypertrophy with accumulation of early germ cells in both sexes, namely spermatogonia in the testis and primary growth oocytes in the ovary. In amh mutant females, the folliculogenesis was normal in young fish but receded progressively in adults, which was accompanied by progressive decrease in follicle-stimulating hormone (fshb) expression. Interestingly the expression of fshb increased in the pituitary of juvenile amh mutant males but decreased in adults. The upregulation of fshb in mutant male juveniles was likely one of the mechanisms for triggering gonadal hypergrowth, whereas the downregulation of fshb in adults might involve a negative feedback by gonadal inhibin. Further analysis using mutants of fshb and growth differentiation factor 9 (gdf9) provided evidence for a role of FSH in triggering ovarian hypertrophy in young female amh mutant as well. In summary, the present study provided comprehensive genetic evidence for dual roles of Amh in controlling zebrafish gonadal homeostasis and gametogenesis in both sexes. Amh suppresses proliferation or accumulation of early germ cells (spermatogonia in testis and primary growth oocytes in ovary) while promoting their exit to advanced stages, and its action may involve both endocrine and paracrine pathways.
Collapse
Affiliation(s)
- Zhiwei Zhang
- Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau
| | - Bo Zhu
- Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau
| | - Weiting Chen
- Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau
| | - Wei Ge
- Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau.
| |
Collapse
|
17
|
Nyuji M, Hongo Y, Yoneda M, Nakamura M. Transcriptome characterization of BPG axis and expression profiles of ovarian steroidogenesis-related genes in the Japanese sardine. BMC Genomics 2020; 21:668. [PMID: 32993516 PMCID: PMC7526130 DOI: 10.1186/s12864-020-07080-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 09/18/2020] [Indexed: 11/10/2022] Open
Abstract
Background The clupeoid fishes are ecologically and commercially important fish species worldwide that exhibit a high level of population fluctuation, accompanied by alteration of reproductive traits. However, knowledge about their reproductive physiology in order to understand mechanisms underlying such population dynamics is limited. The endocrine system along with the brain–pituitary–gonadal (BPG) axis is critical for regulating reproduction. The aims of this study were to provide transcript data and genes related to the BPG axis, and to characterize the expression profiles of ovarian steroidogenesis-related genes in the Japanese sardine (Sardinops melanostictus, Clupeidae). Results RNA sequencing was performed using the sardine brain, pituitary, and gonad in both sexes. A total of 290,119 contigs were obtained and 115,173 non-redundant ORFs were annotated. The genes differentially expressed between ovary and testis were strongly associated with GO terms related to gamete production. The tissue-specific profile of the abundance of transcripts was characterized for the major regulators in the BPG axis, such as gonadotropin-releasing hormone, gonadotropin, and steroidogenic enzyme. By comparing between ovary and testis, out of eight different 17β-hydroxysteroid dehydrogenase (Hsd17b) genes identified, higher hsd17b7 expression was found in testis, whereas higher expression of hsd17b8, hsd17b10, hsd17b12a, and hsd17b12b was found in ovary. The cDNAs encoding key endocrine factors in the ovarian steroidogenic pathway were cloned, sequenced, and quantitatively assayed. In the pituitary, follicle-stimulating hormone beta peaked during vitellogenesis, while luteinizing hormone beta peaked at the completion of vitellogenesis. In the ovary, follicle-stimulating hormone receptor and luteinizing hormone receptor were upregulated from mid- to late phase of vitellogenesis. Furthermore, three steroidogenic enzyme genes (cyp11a1, cyp17a1, and cyp19a1a) gradually increased their expression during ovarian development, accompanying a rise in serum estradiol-17β, while 3β-hydroxysteroid dehydrogenase and steroidogenic acute regulatory protein did not change significantly. Conclusions This is the first report of deep RNA sequencing analysis of Japanese sardine, in which many key genes involved in the BPG axis were identified. Expression profiles of ovarian steroidogenesis-related genes provide a molecular basis of the physiological processes underlying ovarian development in the sardine. Our study will be a valuable resource for clarifying the molecular biology of clupeoid fishes.
Collapse
Affiliation(s)
- Mitsuo Nyuji
- Fisheries Resources Institute, Japan Fisheries Research and Education Agency, Yokohama, 236-8648, Japan.
| | - Yuki Hongo
- Fisheries Resources Institute, Japan Fisheries Research and Education Agency, Yokohama, 236-8648, Japan
| | - Michio Yoneda
- Hakatajima Field Station, Fisheries Technology Institute, Japan Fisheries Research and Education Agency, Kinoura, Imabari, Ehime, 794-2305, Japan
| | - Masahiro Nakamura
- Hakatajima Field Station, Fisheries Technology Institute, Japan Fisheries Research and Education Agency, Kinoura, Imabari, Ehime, 794-2305, Japan
| |
Collapse
|
18
|
Peter Dennis L, Nocillado J, Palma P, Amagai T, Soyano K, Elizur A. Development of a giant grouper Luteinizing Hormone (LH) Enzyme-Linked Immunosorbent Assay (ELISA) and its use towards understanding sexual development in grouper. Gen Comp Endocrinol 2020; 296:113542. [PMID: 32585213 DOI: 10.1016/j.ygcen.2020.113542] [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: 02/28/2020] [Revised: 06/06/2020] [Accepted: 06/16/2020] [Indexed: 10/23/2022]
Abstract
A recombinant giant grouper Luteinizing Hormone (LH) consisting of tethered beta and alpha subunits was produced in a yeast expression system. The giant grouper LH β-subunit was also produced and administered to rabbits for antibody development. The recombinant LH and its antibody were used to develop an Enzyme Linked Immunosorbent Assay (ELISA). This ELISA enabled detection of plasma LH levels in groupers at a sensitivity between 391 pg/ml and 200 ng/ml. Different species of grouper were assayed with this ELISA in conjunction with gonadal histology and body condition data to identify links between circulating LH levels and sexual development. We found that circulating levels of LH decreased when oocytes began to degenerate, and sex-transition gonadal characteristics were apparent when LH levels decreased further. When circulating LH levels were related to body condition (body weight/ body length), transitioning-stage fish had relatively high body condition but low plasma LH levels. This observation was similar across multiple grouper species and indicates that plasma LH levels combined with body condition may be a marker for early male identification in the protogynous hermaphrodite groupers.
Collapse
Affiliation(s)
- Lachlan Peter Dennis
- Genecology Research Centre, University of the Sunshine Coast, Locked Bag 4, Maroochydore DC 4558, Queensland, Australia
| | - Josephine Nocillado
- Genecology Research Centre, University of the Sunshine Coast, Locked Bag 4, Maroochydore DC 4558, Queensland, Australia
| | - Peter Palma
- Genecology Research Centre, University of the Sunshine Coast, Locked Bag 4, Maroochydore DC 4558, Queensland, Australia; Aquaculture Department, Southeast Asian Fisheries Development Center, Tigbauan 5021, Iloilo, Philippines
| | - Takafumi Amagai
- Institute for East China Sea Research, Nagasaki University, Bunkyomachi 852-8131, Nagasaki, Japan
| | - Kiyoshi Soyano
- Institute for East China Sea Research, Nagasaki University, Bunkyomachi 852-8131, Nagasaki, Japan
| | - Abigail Elizur
- Genecology Research Centre, University of the Sunshine Coast, Locked Bag 4, Maroochydore DC 4558, Queensland, Australia.
| |
Collapse
|
19
|
Koua ND, Núñez-Rodriguez J, Orjuela J, Zatylny-Gaudin C, Dubos MP, Bernay B, Pontin J, Corre E, Henry J. Identification and structural characterization of the factors involved in vitellogenesis and its regulation in the African Osteoglossiforme of aquacultural interest Heterotis niloticus (Cuvier, 1829). Gen Comp Endocrinol 2020; 296:113532. [PMID: 32535172 DOI: 10.1016/j.ygcen.2020.113532] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/29/2020] [Accepted: 06/03/2020] [Indexed: 11/25/2022]
Abstract
The African bonytongue (Heterotis niloticus) is an excellent candidate for fish farming because it has outstanding biological characteristics and zootechnical performances. However, the absence of sexual dimorphism does not favor its reproduction in captivity or the understanding of its reproductive behavior. Moreover, no molecular data related to its reproduction is yet available. This study therefore focuses on the structural identification of the different molecular actors of vitellogenesis expressed in the pituitary gland, the liver and the ovary of H. niloticus. A transcriptomic approach based on de novo RNA sequencing of the pituitary gland, ovary and liver of females in vitellogenesis led to the creation of three transcriptomes. In silico analysis of these transcriptomes identified the sequences of pituitary hormones such as prolactin (PRL), luteinizing hormone (LH) and follicle-stimulating hormone (FSH) and their ovarian receptors (PRLR, FSHR, LHR). In the liver and ovary, estrogen receptors (ER) beta and gamma, liver vitellogenins (VtgB and VtgC) and their ovarian receptors (VLDLR) were identified. Finally, the partial transcript of an ovarian Vtg weakly expressed compared to hepatic Vtg was identified based on structural criteria. Moreover, a proteomic approach carried out from mucus revealed the presence of one Vtg exclusively in females in vitellogenesis. In this teleost fish that does not exhibit sexual dimorphism, mucus Vtg could be used as a sexing biomarker based on a non-invasive technique compatible with the implementation of experimental protocols in vivo.
Collapse
Affiliation(s)
- N'Zi Daniel Koua
- NORMANDIE UNIV, UNICAEN, CNRS, BOREA, 14000 Caen, France; INP-HB, Département FOREN, BP 1313 Yamoussoukro, Cote d'Ivoire; Laboratoire de Biologie des Organismes et Ecosystèmes Aquatiques (BOREA), Université de Caen-Normandie, MNHN, SU, UA, CNRS, IRD, Esplanade de la paix, 14032 Caen Cedex, France
| | | | | | - Céline Zatylny-Gaudin
- NORMANDIE UNIV, UNICAEN, CNRS, BOREA, 14000 Caen, France; Laboratoire de Biologie des Organismes et Ecosystèmes Aquatiques (BOREA), Université de Caen-Normandie, MNHN, SU, UA, CNRS, IRD, Esplanade de la paix, 14032 Caen Cedex, France
| | - Marie-Pierre Dubos
- NORMANDIE UNIV, UNICAEN, CNRS, BOREA, 14000 Caen, France; Laboratoire de Biologie des Organismes et Ecosystèmes Aquatiques (BOREA), Université de Caen-Normandie, MNHN, SU, UA, CNRS, IRD, Esplanade de la paix, 14032 Caen Cedex, France
| | - Benoît Bernay
- NORMANDIE UNIV, UNICAEN, SF ICORE, Proteogen Platform, Esplanade de la paix, 14032 Caen, France
| | - Julien Pontin
- NORMANDIE UNIV, UNICAEN, SF ICORE, Proteogen Platform, Esplanade de la paix, 14032 Caen, France
| | - Erwan Corre
- Sorbonne Université, CNRS, FR2424, ABiMS, Station Biologique, F-29680 Roscoff, France
| | - Joël Henry
- NORMANDIE UNIV, UNICAEN, CNRS, BOREA, 14000 Caen, France; Laboratoire de Biologie des Organismes et Ecosystèmes Aquatiques (BOREA), Université de Caen-Normandie, MNHN, SU, UA, CNRS, IRD, Esplanade de la paix, 14032 Caen Cedex, France.
| |
Collapse
|
20
|
Song Y, Chen J, Tao B, Luo D, Zhu Z, Hu W. Kisspeptin2 regulates hormone expression in female zebrafish (Danio rerio) pituitary. Mol Cell Endocrinol 2020; 513:110858. [PMID: 32413385 DOI: 10.1016/j.mce.2020.110858] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 04/09/2020] [Accepted: 04/30/2020] [Indexed: 01/01/2023]
Abstract
Kisspeptin2 is a neuropeptide widely found in the brain and multiple peripheral tissues in the zebrafish. The pituitary is the center of synthesis and secretes various endocrine hormones. However, Kiss2 innervation in the zebrafish pituitary is unknown. In this study, the organization of Kiss2 cells and structures in the zebrafish pituitary by promoter-driving mCherry-labeling Kiss2 neurons were investigated. Kiss2 neurons in the hypothalamus do not project into the pituitary. Kiss2 cells are found in the female pituitary. Unidentified Kiss2 cells and extensions are located in the proximal pars distalis (PPD), similar to the distribution of Gnrh3 fibers. Kiss2 structures reside alongside Gnrh3 fibers. No Kiss2 structures are found in the male pituitary. The transcriptional expression of the kisspeptin receptor kiss1rb is detected in both female and male pituitaries. In situ hybridization shows that kiss1rb-positive cells are located in the PPD and pars intermedia (PI). In vitro Kiss2-10 treatment stimulates Akt and Erk phosphorylation and significantly induces lhβ, fshβ, and prl1 mRNA expression in the female pituitary. The results in this study suggest that Kiss2 and Kiss1rb may form an independent paracrine or autocrine system in the female zebrafish pituitary. Kiss2 and Kiss1rb signaling regulates the expression of pituitary hormones.
Collapse
Affiliation(s)
- Yanlong Song
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Ji Chen
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Binbin Tao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Daji Luo
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Zuoyan Zhu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Wei Hu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China; Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China.
| |
Collapse
|
21
|
Li J, Ge W. Zebrafish as a model for studying ovarian development: Recent advances from targeted gene knockout studies. Mol Cell Endocrinol 2020; 507:110778. [PMID: 32142861 DOI: 10.1016/j.mce.2020.110778] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 02/27/2020] [Accepted: 02/27/2020] [Indexed: 12/11/2022]
Abstract
Ovarian development is a complex process controlled by precise coordination of multiple factors. The targeted gene knockout technique is a powerful tool to study the functions of these factors. The successful application of this technique in mice in the past three decades has significantly enhanced our understanding on the molecular mechanism of ovarian development. Recently, with the advent of genome editing techniques, targeted gene knockout research can be carried out in many species. Zebrafish has emerged as an excellent model system to study the control of ovarian development. Dozens of genes related to ovarian development have been knocked out in zebrafish in recent years. Much new information and perspectives on the molecular mechanism of ovarian development have been obtained from these mutant zebrafish. Some findings have challenged conventional views. Several genes have been identified for the first time in vertebrates to control ovarian development. Focusing on ovarian development, the purpose of this review is to briefly summarize recent findings using these gene knockout zebrafish models, and compare these findings with mammalian models. These established mutants and rapid development of gene knockout techniques have prompted zebrafish as an ideal animal model for studying ovarian development.
Collapse
Affiliation(s)
- Jianzhen Li
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu, China, 730070.
| | - Wei Ge
- Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau, China.
| |
Collapse
|
22
|
Imamura S, Hur SP, Takeuchi Y, Badruzzaman M, Mahardini A, Rizky D, Takemura A. The mRNA expression patterns of kisspeptins, GnRHs, and gonadotropins in the brain and pituitary gland of a tropical damselfish, Chrysiptera cyanea, during the reproductive cycle. FISH PHYSIOLOGY AND BIOCHEMISTRY 2020; 46:277-291. [PMID: 31705423 DOI: 10.1007/s10695-019-00715-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 10/09/2019] [Indexed: 06/10/2023]
Abstract
The sapphire devil (Chrysiptera cyanea) is a tropical damselfish that undergoes active reproduction under long-day conditions. To elucidate the physiological regulation of the brain-pituitary-gonadal axis in female sapphire devil, we cloned and characterized the genes of two kisspeptins (kiss1 and kiss2), three gonadotropin-releasing hormones (gnrh1, gnrh2, gnrh3), and the β-subunit of two gonadotropins (fshβ and lhβ) and investigated the gene expression changes during ovarian development. Quantitative polymerase chain reaction analyses in various brain parts revealed high expression levels of kiss1, kiss2, and gnrh2 in the diencephalon; gnrh2 and gnrh3 in the telencephalon; and fshβ and lhβ in the pituitary. In situ hybridization (ISH) analyses revealed positive signals of kiss1 in the dorsal and ventral habenular nucleus and of kiss2 in the dorsal and ventral parts of the nucleus of the lateral recess. This analysis showed gnrh1 expression in the preoptic area (POA), suggesting that GnRH1 plays a stimulating role in the secretion of gonadotropins from the pituitary of the sapphire devil. High transcription levels of kiss1, kiss2, gnrh1, gnrh2, fshβ, and lhβ were observed in the brain during the late vitellogenic stage, suggesting their involvement in the physiological processes of vitellogenesis. Immersion of fish in estradiol-17β (E2)-containing seawater resulted in increased expression of kiss2 and gnrh1 in their brains. This study showed that kiss-expressing neurons in the diencephalon are influenced by E2, leading to upregulation of gnrh1 in the POA and of fshβ and lhβ in the pituitary during vitellogenesis.
Collapse
Affiliation(s)
- Satoshi Imamura
- Department of Chemistry, Biology and Marine Science, Faculty of Science, University of the Ryukyus, Senbaru 1, Nishihara, Okinawa, 903-0213, Japan
| | - Sung-Pyo Hur
- Jeju Research Institute, Korea Institute of Ocean Science & Technology, Jeju, 63349, South Korea
| | - Yuki Takeuchi
- Department of Chemistry, Biology and Marine Science, Faculty of Science, University of the Ryukyus, Senbaru 1, Nishihara, Okinawa, 903-0213, Japan
- Okinawa Institute of Science and Technology Graduate School, 1919-1 Tancha, Onna, Okinawa, 904-0495, Japan
| | - Muhammad Badruzzaman
- Department of Chemistry, Biology and Marine Science, Faculty of Science, University of the Ryukyus, Senbaru 1, Nishihara, Okinawa, 903-0213, Japan
- Department of Biochemistry and Molecular Biology, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Salna, Gazipur, 1706, Bangladesh
| | - Angka Mahardini
- Department of Chemistry, Biology and Marine Science, Faculty of Science, University of the Ryukyus, Senbaru 1, Nishihara, Okinawa, 903-0213, Japan
| | - Dinda Rizky
- Department of Chemistry, Biology and Marine Science, Faculty of Science, University of the Ryukyus, Senbaru 1, Nishihara, Okinawa, 903-0213, Japan
| | - Akihiro Takemura
- Department of Chemistry, Biology and Marine Science, Faculty of Science, University of the Ryukyus, Senbaru 1, Nishihara, Okinawa, 903-0213, Japan.
| |
Collapse
|
23
|
Burow S, Mizrahi N, Maugars G, von Krogh K, Nourizadeh-Lillabadi R, Hollander-Cohen L, Shpilman M, Atre I, Weltzien FA, Levavi-Sivan B. Characterization of gonadotropin receptors Fshr and Lhr in Japanese medaka, Oryzias latipes. Gen Comp Endocrinol 2020; 285:113276. [PMID: 31536722 DOI: 10.1016/j.ygcen.2019.113276] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 09/14/2019] [Accepted: 09/14/2019] [Indexed: 10/26/2022]
Abstract
Reproduction in vertebrates is controlled by the brain-pituitary-gonad axis, where the two gonadotropins follicle-stimulating hormone (Fsh) and luteinizing hormone (Lh) play vital parts by activating their cognate receptors in the gonads. The main purpose of this work was to study intra- and interspecies ligand promiscuity of teleost gonadotropin receptors, since teleost receptor specificity is unclear, in contrast to mammalian receptors. Receptor activation was investigated by transfecting COS-7 cells with either Fsh receptor (mdFshr, tiFshr) or Lh receptor (mdLhr, tiLhr), and tested for activation by recombinant homologous and heterologous ligands (mdFshβα, mdLhβα, tiFshβα, tiLhβα) from two representative fish orders, Japanese medaka (Oryzias latipes, Beloniformes) and Nile tilapia (Oreochromis niloticus, Cichliformes). Results showed that each gonadotropin preferentially activates its own cognate receptor. Cross-reactivity was detected to some extent as mdFshβα was able to activate the mdLhr, and mdLhβα the mdFshr. Medaka pituitary extract (MPE) stimulated CRE-LUC activity in COS-7 cells expressing mdlhr, but could not stimulate cells expressing mdfshr. Recombinant tiLhβα, tiFshβα and tilapia pituitary extract (TPE) could activate the mdLhr, suggesting cross-species reactivity for mdLhr. Cross-species reactivity was also detected for mdFshr due to activation by tiFshβα, tiLhβα, and TPE, as well as for tiFshr and tiLhr due to stimulation by mdFshβα, mdLhβα, and MPE. Tissue distribution analysis of gene expression revealed that medaka receptors, fshr and lhr, are highly expressed in both ovary and testis. High expression levels were found for lhr also in brain, while fshr was expressed at low levels. Both fshr and lhr mRNA levels increased significantly during testis development. Amino acid sequence alignment and three-dimensional modelling of ligands and receptors highlighted conserved beta sheet domains of both Fsh and Lh between Japanese medaka and Nile tilapia. It also showed a higher structural homology and similarity of transmembrane regions of Lhr between both species, in contrast to Fshr, possibly related to the substitution of the conserved cysteine residue in the transmembrane domain 6 in medaka Fshr with glycine. Taken together, this is the first characterization of medaka Fshr and Lhr using homologous ligands, enabling to better understand teleost hormone-receptor interactions and specificities. The data suggest partial ligand promiscuity and cross-species reactivity between gonadotropins and their receptors in medaka and tilapia.
Collapse
Affiliation(s)
- Susann Burow
- Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, 0454 Oslo, Norway
| | - Naama Mizrahi
- Department of Animal Sciences, Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot 76100, Israel
| | - Gersende Maugars
- Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, 0454 Oslo, Norway
| | - Kristine von Krogh
- Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, 0454 Oslo, Norway
| | - Rasoul Nourizadeh-Lillabadi
- Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, 0454 Oslo, Norway
| | - Lian Hollander-Cohen
- Department of Animal Sciences, Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot 76100, Israel
| | - Michal Shpilman
- Department of Animal Sciences, Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot 76100, Israel
| | - Ishwar Atre
- Department of Animal Sciences, Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot 76100, Israel
| | - Finn-Arne Weltzien
- Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, 0454 Oslo, Norway.
| | - Berta Levavi-Sivan
- Department of Animal Sciences, Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot 76100, Israel.
| |
Collapse
|
24
|
Liu DT, Hong WS, Chen SX, Zhu Y. Upregulation of adamts9 by gonadotropin in preovulatory follicles of zebrafish. Mol Cell Endocrinol 2020; 499:110608. [PMID: 31586455 PMCID: PMC6878983 DOI: 10.1016/j.mce.2019.110608] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 09/30/2019] [Accepted: 10/01/2019] [Indexed: 02/05/2023]
Abstract
Previously we had identified adamts9 as a downstream target of Pgr, which is essential for ovulation in zebrafish. The primary goal of this study is to determine whether human chorionic gonadotropin (hCG, LH analog) also regulate adamts9 expression prior to ovulation. The expression of adamts9 was induced by hCG in a dose and time dependent manner in zebrafish preovulatory follicles in vitro. Interestingly, the stimulatory effect of hCG on adamts9 expression was not blocked in pgr-/- follicles but blocked in lhcgr-/-. This effect of hCG was via Lhcgr and its associated cAMP and PKC signaling pathways. Reduced fecundity and reduced expression of adamts9 were also found in lhcgr-/- females in vivo. Therefore, we have provided the first evidence of gonadotropin (hCG) regulated adamts9 in zebrafish, which could be important for ovulation.
Collapse
Affiliation(s)
- Dong Teng Liu
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, Fujian Province, 361102, People's Republic of China
| | - Wan Shu Hong
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, Fujian Province, 361102, People's Republic of China
| | - Shi Xi Chen
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, Fujian Province, 361102, People's Republic of China.
| | - Yong Zhu
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, Fujian Province, 361102, People's Republic of China; Department of Biology, East Carolina University, 1000 5th Street, Greenville, NC, 27858, USA.
| |
Collapse
|
25
|
Irob K, Wagler M, Baberschke N, Meinelt T, Kloas W. Potash mining effluents induce moderate effects on histopathological and physiological endpoints of adult zebrafish (Danio rerio). THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 694:133471. [PMID: 31400679 DOI: 10.1016/j.scitotenv.2019.07.277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 07/12/2019] [Accepted: 07/17/2019] [Indexed: 06/10/2023]
Abstract
Stress in fish can be caused by a variety of factors and has the potential to evoke stress responses leading to a reduction of physical condition and of health. The river Werra (Germany) presents a severe case of secondary salinisation caused by potash mining activities. The model organism Danio rerio was exposed to different ion-concentrations depicting current (HT) and future (LT) threshold values of the Werra, as well as to solutions with single-exceeding ions (Mg2+ + K+ (KMg), Mg2+ (Mg) and K+ (K)). After a six-week exposure period, cortisol levels, growth and weight were measured, gills and gonads were histologically analysed and mRNA expression of follicle stimulating hormone (FSH), luteinising hormone (LH), growth hormone (GH) and prolactin (PRL) were determined. Cortisol was still elevated in fish in the HT and K group, indicating moderate stress. However, gills revealed structural changes in zebrafish in all exposure groups, size of oocytes differed in the LT and K group, male FSH mRNA levels were elevated in the HT and LT group whereas PRL mRNA levels were lower in HT and LT for both, male and female fish. These results suggest that ion-stress induces moderate effects on a variety of biological parameters that mainly serve to adapt to elevated ion concentrations. For these reasons current and even future thresholds should be reconsidered, including thresholds for total as well as single ion concentrations. Future research looking at the effects on local fish species is needed, along with regular and long-term monitoring of environmental conditions, species abundance and diversity.
Collapse
Affiliation(s)
- Katja Irob
- Department of Ecophysiology and Aquaculture, Leibniz Institute of Freshwater Ecology and Inland Fisheries, Müggelseedamm 310, Berlin 12587, Germany; Biodiversity and Theoretical Ecology, Institute of Biology, Freie Universität Berlin, Altensteinstr. 34, Berlin 14195, Germany.
| | - Marit Wagler
- Department of Ecophysiology and Aquaculture, Leibniz Institute of Freshwater Ecology and Inland Fisheries, Müggelseedamm 310, Berlin 12587, Germany
| | - Nora Baberschke
- Department of Ecophysiology and Aquaculture, Leibniz Institute of Freshwater Ecology and Inland Fisheries, Müggelseedamm 310, Berlin 12587, Germany
| | - Thomas Meinelt
- Department of Ecophysiology and Aquaculture, Leibniz Institute of Freshwater Ecology and Inland Fisheries, Müggelseedamm 310, Berlin 12587, Germany
| | - Werner Kloas
- Department of Ecophysiology and Aquaculture, Leibniz Institute of Freshwater Ecology and Inland Fisheries, Müggelseedamm 310, Berlin 12587, Germany; Department of Endocrinology, Institute of Biology and Albrecht Daniel Thaer-Institute, Faculty of Life Sciences, Humboldt Universität zu Berlin, Invalidenstr. 42, Berlin 10115, Germany
| |
Collapse
|
26
|
Kanda S. Evolution of the regulatory mechanisms for the hypothalamic-pituitary-gonadal axis in vertebrates-hypothesis from a comparative view. Gen Comp Endocrinol 2019; 284:113075. [PMID: 30500374 DOI: 10.1016/j.ygcen.2018.11.014] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 11/26/2018] [Accepted: 11/26/2018] [Indexed: 12/26/2022]
Abstract
Reproduction is regulated by the hypothalamic-pituitary-gonadal (HPG) axis in vertebrates. In addition to wealth of knowledge in mammals, recent studies in non-mammalian species, especially teleosts, have provided evidence that some of the components in the HPG axis are conserved in bony vertebrates. On the other hand, from the comparisons of the recent accumulating knowledge between mammals and teleosts, unique characteristics of the regulatory system in each group have been unveiled. A hypophysiotropic neurotransmitter/hormone, gonadotropin releasing hormone (GnRH), pituitary gonadotropins, follicle stimulating hormone (FSH), and luteinizing hormone (LH) were proven to be common important elements of the HPG axis in teleosts and mammals, although the roles of each vary. Conversely, there are some modulators of GnRH or gonadotropins that are not common to all vertebrates. In this review, I will introduce the mechanism for HPG axis regulation in mammals and teleosts, and describe their evolution from a hypothetical common ancestor.
Collapse
Affiliation(s)
- Shinji Kanda
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo 113-0033, Japan.
| |
Collapse
|
27
|
Safian D, Bogerd J, Schulz RW. Regulation of spermatogonial development by Fsh: The complementary roles of locally produced Igf and Wnt signaling molecules in adult zebrafish testis. Gen Comp Endocrinol 2019; 284:113244. [PMID: 31415728 DOI: 10.1016/j.ygcen.2019.113244] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/07/2019] [Accepted: 08/09/2019] [Indexed: 12/28/2022]
Abstract
Spermatogenesis is a cellular developmental process characterized by the coordinated proliferation and differentiation activities of somatic and germ cells in order to produce a large number of spermatozoa, the cellular basis of male fertility. Somatic cells in the testis, such as Leydig, peritubular myoid and Sertoli cells, provide structural and metabolic support and contribute to the regulatory microenvironment required for proper germ cell survival and development. The pituitary follicle-stimulating hormone (Fsh) is a major endocrine regulator of vertebrate spermatogenesis, targeting somatic cell functions in the testes. In fish, Fsh regulates Leydig and Sertoli cell functions, such as sex steroid and growth factor production, processes that also control the development of spermatogonia, the germ cell stages at the basis of the spermatogenic process. Here, we summarize recent advances in our understanding of mechanisms used by Fsh to regulate the development of spermatogonia. This involves discussing the roles of insulin-like growth factor (Igf) 3 and canonical and non-canonical Wnt signaling pathways. We will also discuss how these locally active regulatory systems interact to maintain testis tissue homeostasis.
Collapse
Affiliation(s)
- Diego Safian
- Reproductive Biology Group, Division Developmental Biology, Institute of Biodynamics and Biocomplexity, Department of Biology, Faculty of Science, University of Utrecht, 3584 CH Utrecht, The Netherlands
| | - Jan Bogerd
- Reproductive Biology Group, Division Developmental Biology, Institute of Biodynamics and Biocomplexity, Department of Biology, Faculty of Science, University of Utrecht, 3584 CH Utrecht, The Netherlands
| | - Rüdiger W Schulz
- Reproductive Biology Group, Division Developmental Biology, Institute of Biodynamics and Biocomplexity, Department of Biology, Faculty of Science, University of Utrecht, 3584 CH Utrecht, The Netherlands; Reproduction and Developmental Biology Group, Institute of Marine Research, P.O. Box 1870, Nordnes, 5817 Bergen, Norway.
| |
Collapse
|
28
|
Hollander-Cohen L, Böhm B, Hausken K, Levavi-Sivan B. Ontogeny of the specificity of gonadotropin receptors and gene expression in carp. Endocr Connect 2019; 8:1433-1446. [PMID: 31581128 PMCID: PMC6826172 DOI: 10.1530/ec-19-0389] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 10/02/2019] [Indexed: 02/02/2023]
Abstract
The pituitary gonadotropins, luteinizing hormone (LH) and follicle-stimulating hormone (FSH), are the principle endocrine drivers of reproductive processes in the gonads of jawed vertebrates. Canonically, FSH recruits and maintains selected ovarian follicles for maturation and LH induces the stages of germinal vesicle breakdown and ovulation. In mammals, LH and FSH specifically activate cognate G-protein-coupled receptors that affect the proteins involved in steroidogenesis, protein hormone synthesis, and gametogenesis. This dual-gonadotropin model also exists in some fish species, but not in all. In fact, due to their diverse number of species, extended number of ecological niches, and remarkably flexible reproductive strategies, fish are appropriate as models to understand the co-evolution of gonadotropins and their receptors. In this study, we cloned and characterized the expression profile over the final stages of ovarian maturation of carp (Cyprinus carpio) LHCGR and FSHR. Expression of both gonadotropin receptors increased in the later stage of early vitellogenesis, suggesting that both LH and FSH play a role in the development of mature follicles. We additionally tested the activation of cLHCGR and cFSHR using homologous and heterologous recombinant gonadotropins in order to gain insight into an evolutionary model of permissive gonadotropin receptor function. These data suggest that carp (Cyprinus carpio) gonad development and maturation depends on a specific gonadotropin profile that does not reflect the temporally distinct dual-gonadotropin model observed in salmonids or mammals, and that permissive gonadotropin receptor activation is a specific feature of Ostariophysi, not all teleosts.
Collapse
Affiliation(s)
- Lian Hollander-Cohen
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, Hebrew University of Jerusalem, Rehovot, Israel
| | - Benjamin Böhm
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, Hebrew University of Jerusalem, Rehovot, Israel
| | - Krist Hausken
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, Hebrew University of Jerusalem, Rehovot, Israel
| | - Berta Levavi-Sivan
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, Hebrew University of Jerusalem, Rehovot, Israel
- Correspondence should be addressed to B Levavi-Sivan:
| |
Collapse
|
29
|
Dufour S, Quérat B, Tostivint H, Pasqualini C, Vaudry H, Rousseau K. Origin and Evolution of the Neuroendocrine Control of Reproduction in Vertebrates, With Special Focus on Genome and Gene Duplications. Physiol Rev 2019; 100:869-943. [PMID: 31625459 DOI: 10.1152/physrev.00009.2019] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In humans, as in the other mammals, the neuroendocrine control of reproduction is ensured by the brain-pituitary gonadotropic axis. Multiple internal and environmental cues are integrated via brain neuronal networks, ultimately leading to the modulation of the activity of gonadotropin-releasing hormone (GnRH) neurons. The decapeptide GnRH is released into the hypothalamic-hypophysial portal blood system and stimulates the production of pituitary glycoprotein hormones, the two gonadotropins luteinizing hormone and follicle-stimulating hormone. A novel actor, the neuropeptide kisspeptin, acting upstream of GnRH, has attracted increasing attention in recent years. Other neuropeptides, such as gonadotropin-inhibiting hormone/RF-amide related peptide, and other members of the RF-amide peptide superfamily, as well as various nonpeptidic neuromediators such as dopamine and serotonin also provide a large panel of stimulatory or inhibitory regulators. This paper addresses the origin and evolution of the vertebrate gonadotropic axis. Brain-pituitary neuroendocrine axes are typical of vertebrates, the pituitary gland, mediator and amplifier of brain control on peripheral organs, being a vertebrate innovation. The paper reviews, from molecular and functional perspectives, the evolution across vertebrate radiation of some key actors of the vertebrate neuroendocrine control of reproduction and traces back their origin along the vertebrate lineage and in other metazoa before the emergence of vertebrates. A focus is given on how gene duplications, resulting from either local events or from whole genome duplication events, and followed by paralogous gene loss or conservation, might have shaped the evolutionary scenarios of current families of key actors of the gonadotropic axis.
Collapse
Affiliation(s)
- Sylvie Dufour
- Muséum National d'Histoire Naturelle, Biology of Aquatic Organisms and Ecosystems, CNRS, IRD, Sorbonne Université, Université Caen Normandie, Université des Antilles, Paris, France; Université Paris Diderot, Sorbonne Paris Cite, Biologie Fonctionnelle et Adaptative, Paris, France; INSERM U1133, Physiologie de l'axe Gonadotrope, Paris, France; Muséum National d'Histoire Naturelle, Physiologie Moléculaire et Adaptation, Muséum National d'Histoire Naturelle, Paris, France; Université Paris-Saclay, Université Paris-Sud, CNRS, Paris-Saclay Institute of Neuroscience (UMR 9197), Gif-sur-Yvette, France; and Université de Rouen Normandie, Rouen, France
| | - Bruno Quérat
- Muséum National d'Histoire Naturelle, Biology of Aquatic Organisms and Ecosystems, CNRS, IRD, Sorbonne Université, Université Caen Normandie, Université des Antilles, Paris, France; Université Paris Diderot, Sorbonne Paris Cite, Biologie Fonctionnelle et Adaptative, Paris, France; INSERM U1133, Physiologie de l'axe Gonadotrope, Paris, France; Muséum National d'Histoire Naturelle, Physiologie Moléculaire et Adaptation, Muséum National d'Histoire Naturelle, Paris, France; Université Paris-Saclay, Université Paris-Sud, CNRS, Paris-Saclay Institute of Neuroscience (UMR 9197), Gif-sur-Yvette, France; and Université de Rouen Normandie, Rouen, France
| | - Hervé Tostivint
- Muséum National d'Histoire Naturelle, Biology of Aquatic Organisms and Ecosystems, CNRS, IRD, Sorbonne Université, Université Caen Normandie, Université des Antilles, Paris, France; Université Paris Diderot, Sorbonne Paris Cite, Biologie Fonctionnelle et Adaptative, Paris, France; INSERM U1133, Physiologie de l'axe Gonadotrope, Paris, France; Muséum National d'Histoire Naturelle, Physiologie Moléculaire et Adaptation, Muséum National d'Histoire Naturelle, Paris, France; Université Paris-Saclay, Université Paris-Sud, CNRS, Paris-Saclay Institute of Neuroscience (UMR 9197), Gif-sur-Yvette, France; and Université de Rouen Normandie, Rouen, France
| | - Catherine Pasqualini
- Muséum National d'Histoire Naturelle, Biology of Aquatic Organisms and Ecosystems, CNRS, IRD, Sorbonne Université, Université Caen Normandie, Université des Antilles, Paris, France; Université Paris Diderot, Sorbonne Paris Cite, Biologie Fonctionnelle et Adaptative, Paris, France; INSERM U1133, Physiologie de l'axe Gonadotrope, Paris, France; Muséum National d'Histoire Naturelle, Physiologie Moléculaire et Adaptation, Muséum National d'Histoire Naturelle, Paris, France; Université Paris-Saclay, Université Paris-Sud, CNRS, Paris-Saclay Institute of Neuroscience (UMR 9197), Gif-sur-Yvette, France; and Université de Rouen Normandie, Rouen, France
| | - Hubert Vaudry
- Muséum National d'Histoire Naturelle, Biology of Aquatic Organisms and Ecosystems, CNRS, IRD, Sorbonne Université, Université Caen Normandie, Université des Antilles, Paris, France; Université Paris Diderot, Sorbonne Paris Cite, Biologie Fonctionnelle et Adaptative, Paris, France; INSERM U1133, Physiologie de l'axe Gonadotrope, Paris, France; Muséum National d'Histoire Naturelle, Physiologie Moléculaire et Adaptation, Muséum National d'Histoire Naturelle, Paris, France; Université Paris-Saclay, Université Paris-Sud, CNRS, Paris-Saclay Institute of Neuroscience (UMR 9197), Gif-sur-Yvette, France; and Université de Rouen Normandie, Rouen, France
| | - Karine Rousseau
- Muséum National d'Histoire Naturelle, Biology of Aquatic Organisms and Ecosystems, CNRS, IRD, Sorbonne Université, Université Caen Normandie, Université des Antilles, Paris, France; Université Paris Diderot, Sorbonne Paris Cite, Biologie Fonctionnelle et Adaptative, Paris, France; INSERM U1133, Physiologie de l'axe Gonadotrope, Paris, France; Muséum National d'Histoire Naturelle, Physiologie Moléculaire et Adaptation, Muséum National d'Histoire Naturelle, Paris, France; Université Paris-Saclay, Université Paris-Sud, CNRS, Paris-Saclay Institute of Neuroscience (UMR 9197), Gif-sur-Yvette, France; and Université de Rouen Normandie, Rouen, France
| |
Collapse
|
30
|
Gao Y, Jing Q, Huang B, Jia Y. Molecular cloning, characterization, and mRNA expression of gonadotropins during larval development in turbot (Scophthalmus maximus). FISH PHYSIOLOGY AND BIOCHEMISTRY 2019; 45:1697-1707. [PMID: 31098916 DOI: 10.1007/s10695-019-00656-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 05/06/2019] [Indexed: 06/09/2023]
Abstract
Gonadotropins (GtHs) play a pivotal role in regulating the reproductive axis and puberty. In this study, full-length sequences coding for common glycoprotein α subunit (CGα) and luteinizing hormone β (LHβ) were isolated from female turbot (Scophthalmus maximus) pituitary by homology cloning and a strategy based on rapid amplification of cDNA end-polymerase chain reaction. Results showed that the two cDNAs consisted of 669 and 660 nucleotides encoding 129 and 139 amino acids, respectively. CGα and LHβ manifested typical characteristics of glycoprotein hormones, high homologies with the corresponding sequences of available teleosts, and high homology with that of Hippoglossus hippoglossus. CGα, FSHβ, and LHβ mRNAs were abundant in the pituitary, but less expressed in extra-pituitary tissues. The cgα, fshβ, and lhβ were detected at 1-day post-hatching (dph) and peaked simultaneously at early-metamorphosis (22 dph). cgα and fshβ mRNA levels were significantly increased at pre-metamorphosis, peaked in early metamorphosis, and then gradually decreased until metamorphosis was completed. Conversely, lhβ mRNA levels gradually decreased at pre-metamorphosis, dramatically peaked at early metamorphosis, and then decreased during metamorphosis. In addition, the mRNA levels of cgα were significantly higher than those of fshβ and lhβ during turbot larval metamorphic development, whereas no significant difference was found between fshβ and lhβ. These results suggested (i) an early activation of the GtHs system after hatching, which was the highest expression at early metamorphosis, and (ii) FSHβ and LHβ were together involved in the establishment of the reproductive axis during larval development in turbot. These findings contribute to further understanding the potential roles of GtHs during fish larval development.
Collapse
Affiliation(s)
- Yunhong Gao
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, No. 106 Nanjing Road, Qingdao, 266071, People's Republic of China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Qiqi Jing
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, No. 106 Nanjing Road, Qingdao, 266071, People's Republic of China
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Bin Huang
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, No. 106 Nanjing Road, Qingdao, 266071, People's Republic of China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Yudong Jia
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, No. 106 Nanjing Road, Qingdao, 266071, People's Republic of China.
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China.
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China.
| |
Collapse
|
31
|
Schulz RW, Taranger GL, Bogerd J, Nijenhuis W, Norberg B, Male R, Andersson E. Entry into puberty is reflected in changes in hormone production but not in testicular receptor expression in Atlantic salmon (Salmo salar). Reprod Biol Endocrinol 2019; 17:48. [PMID: 31226998 PMCID: PMC6588918 DOI: 10.1186/s12958-019-0493-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 06/14/2019] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Puberty in male Atlantic salmon in aquaculture can start as early as after the first winter in seawater, stunts growth and entails welfare problems due to the maturation-associated loss of osmoregulation capacity in seawater. A better understanding of the regulation of puberty is the basis for developing improved cultivation approaches that avoid these problems. Our aim here was to identify morphological and molecular markers signaling the initiation of, and potential involvement in, testis maturation. METHODS In the first experiment, we monitored for the first time in large Atlantic salmon males several reproductive parameters during 17 months including the first reproductive cycle. Since testicular growth accelerated after the Winter solstice, we focused in the second experiment on the 5 months following the winter solstice, exposing fish from February 1 onwards to the natural photoperiod (NL) or to continuous additional light (LL). RESULTS In the first experiment, testis weight, plasma androgens and pituitary gonadotropin transcript levels increased with the appearance of type B spermatogonia in the testis, but testicular transcript levels for gonadotropin or androgen receptors did not change while being clearly detectable. In the second experiment, all males kept under NL had been recruited into puberty until June. However, recruitment into puberty was blocked in ~ 40% of the males exposed to LL. The first morphological sign of recruitment was an increased proliferation activity of single spermatogonia and Sertoli cells. Irrespective of the photoperiod, this early sign of testis maturation was accompanied by elevated pituitary gnrhr4 and fshb and testicular igf3 transcript levels as well as increased plasma androgen levels. The transition into puberty occurred again with stable testicular gonadotropin and androgen receptor transcript levels. CONCLUSIONS The sensitivity to reproductive hormones is already established before puberty starts and up-regulation of testicular hormone receptor expression is not required to facilitate entry into puberty. The increased availability of receptor ligands, on the other hand, may result from an up-regulation of pituitary Gnrh receptor expression, eventually activating testicular growth factor and sex steroid release and driving germ and Sertoli cell proliferation and differentiation.
Collapse
Affiliation(s)
- Rüdiger W Schulz
- Research Group Reproduction and Developmental Biology, Institute of Marine Research, P.O.Box 1870 Nordnes, 5817, Bergen, Norway
- Reproductive Biology Group, Division Developmental Biology, Department Biology, Science Faculty, Utrecht University, Utrecht, The Netherlands
| | - Geir Lasse Taranger
- Research Group Reproduction and Developmental Biology, Institute of Marine Research, P.O.Box 1870 Nordnes, 5817, Bergen, Norway
| | - Jan Bogerd
- Reproductive Biology Group, Division Developmental Biology, Department Biology, Science Faculty, Utrecht University, Utrecht, The Netherlands
| | - Wouter Nijenhuis
- Reproductive Biology Group, Division Developmental Biology, Department Biology, Science Faculty, Utrecht University, Utrecht, The Netherlands
| | - Birgitta Norberg
- Research Group Reproduction and Developmental Biology, Institute of Marine Research, P.O.Box 1870 Nordnes, 5817, Bergen, Norway
| | - Rune Male
- Department of Biological Sciences, University of Bergen, Bergen, Norway
| | - Eva Andersson
- Research Group Reproduction and Developmental Biology, Institute of Marine Research, P.O.Box 1870 Nordnes, 5817, Bergen, Norway.
| |
Collapse
|
32
|
Lakstygal AM, de Abreu MS, Lifanov DA, Wappler-Guzzetta EA, Serikuly N, Alpsyshov ET, Wang D, Wang M, Tang Z, Yan D, Demin KA, Volgin AD, Amstislavskaya TG, Wang J, Song C, Alekseeva P, Kalueff AV. Zebrafish models of diabetes-related CNS pathogenesis. Prog Neuropsychopharmacol Biol Psychiatry 2019; 92:48-58. [PMID: 30476525 DOI: 10.1016/j.pnpbp.2018.11.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/18/2018] [Accepted: 11/22/2018] [Indexed: 12/12/2022]
Abstract
Diabetes mellitus (DM) is a common metabolic disorder that affects multiple organ systems. DM also affects brain processes, contributing to various CNS disorders, including depression, anxiety and Alzheimer's disease. Despite active research in humans, rodent models and in-vitro systems, the pathogenetic link between DM and brain disorders remains poorly understood. Novel translational models and new model organisms are therefore essential to more fully study the impact of DM on CNS. The zebrafish (Danio rerio) is a powerful novel model species to study metabolic and CNS disorders. Here, we discuss how DM alters brain functions and behavior in zebrafish, and summarize their translational relevance to studying DM-related CNS pathogenesis in humans. We recognize the growing utility of zebrafish models in translational DM research, as they continue to improve our understanding of different brain pathologies associated with DM, and may foster the discovery of drugs that prevent or treat these diseases.
Collapse
Affiliation(s)
- Anton M Lakstygal
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Laboratory of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny, Russia
| | - Murilo S de Abreu
- Bioscience Institute, University of Passo Fundo (UPF), Passo Fundo, RS, Brazil; The International Zebrafish Neuroscience Research Consortium (ZNRC), Slidell, LA, USA
| | - Dmitry A Lifanov
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Laboratory of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny, Russia; School of Pharmacy, Southwest University, Chongqing, China
| | | | - Nazar Serikuly
- School of Pharmacy, Southwest University, Chongqing, China
| | | | - DongMei Wang
- School of Pharmacy, Southwest University, Chongqing, China
| | - MengYao Wang
- School of Pharmacy, Southwest University, Chongqing, China
| | - ZhiChong Tang
- School of Pharmacy, Southwest University, Chongqing, China
| | - DongNi Yan
- School of Pharmacy, Southwest University, Chongqing, China
| | - Konstantin A Demin
- Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Laboratory of Biological Psychiatry, Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Andrey D Volgin
- Scientific Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia
| | | | - JiaJia Wang
- Institute for Marine Drugs and Nutrition, Guangdong Ocean University, Zhanjiang, China; Marine Medicine Development Center, Shenzhen Institute, Guangdong Ocean University, Shenzhen, China
| | - Cai Song
- Institute for Marine Drugs and Nutrition, Guangdong Ocean University, Zhanjiang, China; Marine Medicine Development Center, Shenzhen Institute, Guangdong Ocean University, Shenzhen, China
| | - Polina Alekseeva
- Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
| | - Allan V Kalueff
- School of Pharmacy, Southwest University, Chongqing, China; Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Laboratory of Biological Psychiatry, Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Scientific Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia; Ural Federal University, Ekaterinburg, Russia; Russian Scientific Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny, Russia; ZENEREI Research Center, Slidell, LA, USA.
| |
Collapse
|
33
|
Jia Y, Lei J. Molecular function of gonadotrophins and their receptors in the ovarian development of turbot (Scophthalmus maximus). Gen Comp Endocrinol 2019; 277:17-19. [PMID: 30769011 DOI: 10.1016/j.ygcen.2019.02.014] [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: 08/14/2018] [Revised: 12/14/2018] [Accepted: 02/11/2019] [Indexed: 11/19/2022]
Abstract
Gonadotropins (GtHs) and their receptors (follicle-stimulating hormone receptor, FSHR; luteinizing hormone receptor, LHR) are involved in the regulation of gametogenesis and play important roles during the reproductive cycles in vertebrate species, including fish. This minireview focuses on the molecular characterization and quantification of GtHs (common glycoprotein α subunit CGα, FSHβ, and LHβ) and their receptors (FSHR and LHR) throughout the reproductive cycle of female turbot Scophthalmus maximus. Information about GtHs, FSHR, LHR as well as other ligand-receptors interaction from different teleosts are also included in this review for the implications they may have on the functions of GtHs, FSHR and LHR in the reproductive development of turbot. These findings may enhance our understanding of the physiological roles of the GtHs, FSHR and LHR in controlling of flatfish ovarian development during the reproductive cycle and contributing to the improvement of management strategies for turbots in captivity.
Collapse
Affiliation(s)
- Yudong Jia
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao Key Laboratory for Marine Fish Breeding and Biotechnology, Qingdao 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China.
| | - Jilin Lei
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao Key Laboratory for Marine Fish Breeding and Biotechnology, Qingdao 266071, China
| |
Collapse
|
34
|
Fontaine R, Ager-Wick E, Hodne K, Weltzien FA. Plasticity of Lh cells caused by cell proliferation and recruitment of existing cells. J Endocrinol 2019; 240:361-377. [PMID: 30594119 DOI: 10.1530/joe-18-0412] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 11/30/2018] [Indexed: 01/23/2023]
Abstract
Luteinizing hormone (Lh) and follicle-stimulating hormone (Fsh) control reproduction in vertebrates. Using a transgenic line of medaka, in which green fluorescent protein expression is controlled by the endogenous lhb promotor, we studied development and plasticity of Lh cells, comparing juveniles and adults of both genders. Confocal imaging and 3D reconstruction revealed hypertrophy and hyperplasia of Lh cells in both genders from juvenile to adult stages. We show that Lh cell hyperplasia may be caused by recruitment of existing pituitary cells that start to produce lhb, as evidenced by time lapse recordings of primary pituitary cell cultures, and/or through Lh cell proliferation, demonstrated through a combination of 5-bromo-2'-deoxyuridine incubation experiments and proliferating cell nuclear antigen staining. Proliferating Lh cells do not belong to the classical type of multipotent stem cells, as they do not stain with anti-sox2. Estradiol exposure in vivo increased pituitary cell proliferation, particularly Lh cells, whereas pituitary lhb and gpa expression levels decreased. RNA-seq and in situ hybridization showed that Lh cells express two estrogen receptors, esr1 and esr2b, and the aromatase gene cyp19a1b, suggesting a direct effect of estradiol, and possibly androgens, on Lh cell proliferation. In conclusion, our study reveals a high degree of plasticity in the medaka Lh cell population, resulting from a combination of recruitment and cell proliferation.
Collapse
Affiliation(s)
- Romain Fontaine
- Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Oslo, Norway
| | - Eirill Ager-Wick
- Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Oslo, Norway
| | - Kjetil Hodne
- Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Oslo, Norway
| | - Finn-Arne Weltzien
- Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Oslo, Norway
| |
Collapse
|
35
|
Burow S, Fontaine R, von Krogh K, Mayer I, Nourizadeh-Lillabadi R, Hollander-Cohen L, Cohen Y, Shpilman M, Levavi-Sivan B, Weltzien FA. Medaka follicle-stimulating hormone (Fsh) and luteinizing hormone (Lh): Developmental profiles of pituitary protein and gene expression levels. Gen Comp Endocrinol 2019; 272:93-108. [PMID: 30576646 DOI: 10.1016/j.ygcen.2018.12.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 11/30/2018] [Accepted: 12/16/2018] [Indexed: 02/06/2023]
Abstract
The two gonadotropins follicle-stimulating hormone (Fsh) and luteinizing hormone (Lh) are of particular importance within the hypothalamic-pituitary-gonadal (HPG) axis of vertebrates. In the current study, we demonstrate the production and validation of Japanese medaka (Oryzias latipes) recombinant (md) gonadotropins Fshβ (mdFshβ), Lhβ (mdLhβ), Fshβα (mdFshβα), and Lhβα (mdLhβα) by Pichia pastoris, the generation of specific rabbit antibodies against their respective β subunits, and their use within the development and validation of competitive enzyme-linked immunosorbent assays (ELISAs) for quantification of medaka Fsh and Lh. mdFsh and mdLh were produced as single-chain polypeptides by linking the α subunit with mdFshβ or mdLhβ mature protein coding sequences to produce a "tethered" polypeptide with the β-chain at the N-terminal and the α-chain at the C-terminal. The specificity of the antibodies raised against mdFshβ and mdLhβ was determined by immunofluorescence (IF) for Fshβ and Lhβ on medaka pituitary tissue, while comparison with fluorescence in situ hybridization (FISH) for fshb and lhb mRNA was used for validation. Competitive ELISAs were developed using antibodies against mdFshβ or mdLhβ, and the tethered proteins mdFshβα or mdLhβα for standard curves. The standard curve for the Fsh ELISA ranged from 97.6 pg/ml to 50 ng/ml, and for the Lh ELISA from 12.21 pg/ml to 6.25 ng/ml. The sensitivity of the assays for Fsh and Lh was 44.7 and 70.8 pg/ml, respectively. A profile of pituitary protein levels of medaka Fsh and Lh comparing juveniles with adults showed significant increase of protein amount from juvenile group (body length from 12 mm to 16.5 mm) to adult group (body length from 21 mm to 26.5 mm) for both hormones in male medaka. Comparing these data to a developmental profile of pituitary mRNA expression of medaka fshb and lhb, the mRNA expression of lhb also increased during male maturation and a linear regression analysis revealed a significant increase of lhb expression with increased body length that proposes a linear model. However, fshb mRNA expression did not change significantly during male development and therefore was not correlated with body length. In summary, we have developed and validated homologous ELISA assays for medaka Fsh and Lh based on proteins produced in P. pastoris, assays that will be used to study the functions and regulations of Fsh and Lh in more detail.
Collapse
Affiliation(s)
- Susann Burow
- Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, 0454 Oslo, Norway
| | - Romain Fontaine
- Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, 0454 Oslo, Norway
| | - Kristine von Krogh
- Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, 0454 Oslo, Norway
| | - Ian Mayer
- Department of Production Animal Clinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, 0454 Oslo, Norway
| | - Rasoul Nourizadeh-Lillabadi
- Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, 0454 Oslo, Norway
| | - Lian Hollander-Cohen
- Department of Animal Sciences, Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot 76100, Israel
| | - Yaron Cohen
- Department of Animal Sciences, Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot 76100, Israel
| | - Michal Shpilman
- Department of Animal Sciences, Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot 76100, Israel
| | - Berta Levavi-Sivan
- Department of Animal Sciences, Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot 76100, Israel
| | - Finn-Arne Weltzien
- Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, 0454 Oslo, Norway.
| |
Collapse
|
36
|
Peng C, Xiao L, Chen H, Han Y, Huang M, Zhao M, Li S, Liu Y, Yang Y, Zhang H, Zhang Y, Lin H. Cloning, expression and functional characterization of a novel luteinizing hormone receptor in the orange-spotted grouper, Epinephelus coioides. Gen Comp Endocrinol 2018; 267:90-97. [PMID: 29913168 DOI: 10.1016/j.ygcen.2018.06.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 05/06/2018] [Accepted: 06/15/2018] [Indexed: 01/09/2023]
Abstract
Luteinizing hormone receptor (LHR) plays a critical role in reproduction by mediating LH signaling in the gonad. In this study, we cloned a novel lhr gene from the orange-spotted grouper, named glhr2. The cloned complete open reading frame sequence of glhr2 was 2082 bp in length, encoding a protein of 693 amino acids, sharing approximately 50% amino acid identity with glhr1. glhr1 and glhr2 were primarily expressed in gonad, brain and hypothalamus with low expression in other tissues such as gill, spleen, etc. The expressions of both glhr1 and glhr2 increased during vitellogenesis, while decreased during natural female to male sex change. The two gLHRs both could be activated by equine LH or human chorionic gonadotropin, but not by human follicle stimulating hormone. Both gLHR1 and gLHR2 activation stimulated the expression of cAMP response element driven reporter gene in a dose-dependent manner, while gLHR2 but not gLHR1 also activated serum response element driven reporter gene expression. This was the first study to demonstrate that two active LHRs exist in fish with possible different functional roles.
Collapse
Affiliation(s)
- Cheng Peng
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory for Aquatic Economic Animals and Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, People's Republic of China; Guangdong Institute of Applied Biological Resources, Guangzhou 510260, People's Republic of China
| | - Ling Xiao
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory for Aquatic Economic Animals and Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, People's Republic of China
| | - Huimin Chen
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory for Aquatic Economic Animals and Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, People's Republic of China
| | - Yulong Han
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory for Aquatic Economic Animals and Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, People's Republic of China
| | - Minwei Huang
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory for Aquatic Economic Animals and Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, People's Republic of China
| | - Mi Zhao
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory for Aquatic Economic Animals and Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, People's Republic of China
| | - Shuisheng Li
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory for Aquatic Economic Animals and Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, People's Republic of China.
| | - Yun Liu
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory for Aquatic Economic Animals and Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, People's Republic of China
| | - Yuqing Yang
- Marine Fisheries Development Center of Guangdong Province, Huizhou 516081, People's Republic of China
| | - Haifa Zhang
- Marine Fisheries Development Center of Guangdong Province, Huizhou 516081, People's Republic of China
| | - Yong Zhang
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory for Aquatic Economic Animals and Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, People's Republic of China; Marine Fisheries Development Center of Guangdong Province, Huizhou 516081, People's Republic of China.
| | - Haoran Lin
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory for Aquatic Economic Animals and Guangdong Provincial Engineering Technology Research Center for Healthy Breeding of Important Economic Fish, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, People's Republic of China; Guangdong Institute of Applied Biological Resources, Guangzhou 510260, People's Republic of China; College of Ocean, Hainan University, Haikou 570228, People's Republic of China
| |
Collapse
|
37
|
Szymańska K, Kałafut J, Rivero-Müller A. The gonadotropin system, lessons from animal models and clinical cases. ACTA ACUST UNITED AC 2018; 70:561-587. [PMID: 30264954 DOI: 10.23736/s0026-4784.18.04307-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This review article centers upon family of gonadotropin hormones which consists of two pituitary hormones - follicle-stimulating hormone (FSH) and luteinizing hormone (LH) as well as one non-pituitary hormone - human chorionic gonadotropin (hCG) secreted by placenta, and their receptors. Gonadotropins play an essential role in proper sexual development, puberty, gametogenesis, maintenance of pregnancy and male sexual differentiation during the fetal development. They belong to the family of glycoprotein hormones thus they constitute heterodimeric proteins built of common α subunit and hormone-specific β-subunit. Hitherto, several mutations in genes encoding both gonadotropins and their receptors have been identified in humans. Their occurrence resulted in a number of different phenotypes including delayed puberty, primary amenorrhea, hermaphroditism, infertility and hypogonadism. In order to understand the effects of mutations on the phenotype observed in affected patients, detailed molecular studies are required to map the relationship between the structure and function of gonadotropins and their receptors. Nonetheless, in vitro assays are often insufficient to understand physiology. Therefore, several animal models have been developed to unravel the physiological roles of gonadotropins and their receptors.
Collapse
|
38
|
Hollander-Cohen L, Golan M, Aizen J, Shpilman M, Levavi-Sivan B. Characterization of carp gonadotropins: Structure, annual profile, and carp and zebrafish pituitary topographic organization. Gen Comp Endocrinol 2018; 264:28-38. [PMID: 29183794 DOI: 10.1016/j.ygcen.2017.11.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 11/22/2017] [Accepted: 11/23/2017] [Indexed: 11/26/2022]
Abstract
Two gonadotropins, follicle stimulating hormone (FSH) and luteinizing hormone (LH), are important players in the hypothalamic-pituitary-gonadal axis of vertebrates. In the present work, we describe the construction of recombinant (r) common carp (Cyprinus carpio; c) FSH (rcFSH) and LH (rcLH) using the Pichia pastoris system, the generation of specific antibodies against their respective β subunits, and their use in the development and validation of specific ELISAs. We produced carp rLH and rFSH as single-chain polypeptides, wherein the GTH subunit α was joined with either cLHβ or cFSHβ mature protein-coding sequences to form a fusion gene that encodes a yoked polypeptide, in which the GTH β-subunit forms the N-terminal part and the α-subunit forms the C-terminal part. Competitive ELISAs were developed, using primary antibodies against rcLHβ or rcFSHβ, respectively, and rcLHβα or rcFSHβα for the standard curves. The standard curves for cLH paralleled those of pituitary extracts of the homologous fish and also those of other cyprinids species like the black carp (Mylopharyngodon piceus), goldfish (Carassius auratus), silver carp (Hypophthalmichthys molitrix), and grass carp (Ctenopharyngodon idella). We used the specific antibodies raised against cFSH and cLH to study the specific localization of the different GTH cells in the pituitary of carp and its taxonomic relative species - the zebrafish. Both FSH and LH cells are localized in the center of the proximal pars distalis enveloping both sides of the neurohypophysis. LH cells form a continuous population throughout the PPD, while FSH cells are more loosely distributed throughout the same area and form small aggregations. Marked annual changes were encountered in gonadosomatic index (GSI), follicle diameter, mRNA levels and protein levels of FSH and LH. From September to November, all fish had low GSI, and the ovary contained previtellogenic follicles. From December, the GSI level increased and remained high until March, the follicular diameter reached its maximum in January, where the ovary contained large fully grown follicles. Thereafter, spawning occurred through March and April and ended in May, and GSI level and follicle diameter increased again; and the ovary contained mid-vitellogenic follicles. LH pituitary content and mRNA levels were low at pre- and early vitellogenesis, increasing gradually during this process to reach a peak of LH mRNA levels in mid vitellogenic ovary and a peak of LH content in fully grown ovarian follicles. However, no significant change occurred in FSH pituitary content and mRNA levels in vitellogenic fish and in fish during final maturation stages. A dramatic difference was found in the total content of each gonadotropin in the pituitary, with higher LH than FSH. Moreover, follicle diameter was positively and significantly correlated with LH pituitary content and its transcript levels - but not with the pituitary content or mRNA levels of FSH. Taken together, these results indicate that in carp, LH alone is sufficient to regulate both vitellogenesis and final oocyte maturation while FSH may have another, yet undefined role.
Collapse
Affiliation(s)
- Lian Hollander-Cohen
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - Matan Golan
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - Joseph Aizen
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - Michal Shpilman
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - Berta Levavi-Sivan
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, Hebrew University of Jerusalem, Rehovot 76100, Israel.
| |
Collapse
|
39
|
Pradhan A, Nayak M, Samanta M, Panda RP, Rath SC, Giri SS, Saha A. Gonadotropin receptors of Labeo rohita: Cloning and characterization of full-length cDNAs and their expression analysis during annual reproductive cycle. Gen Comp Endocrinol 2018; 263:21-31. [PMID: 29660307 DOI: 10.1016/j.ygcen.2018.04.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 04/10/2018] [Accepted: 04/12/2018] [Indexed: 12/22/2022]
Abstract
Follicle-stimulating hormone (Fsh) and luteinizing hormone (Lh), secreted from pituitary, stimulate gonadal function by binding to their cognate receptors FSH receptor (FSHR), and LH/choriogonadotropin receptor (LHCGR). Rohu (Labeo rohita) is a commercially important seasonal breeder freshwater fish species, but till date, the regulation of expression of gonadotropins and their receptors gene during different phases of annual reproductive cycle has not been investigated. We envisaged the critical role of these molecules during seasonal gonadal development in this carp species. We cloned full- length cDNAs of fshra and lhcgrba from rohu testis using RACE (Rapid amplification of cDNA ends) and analyzed their expression along with fsh and lh by quantitative real time PCR (qRT-PCR) assay at various gonadal developmental stages of the annual reproductive cycle. Full-length rohu fshra and lhcgrba cDNA encodes 670 and 716 amino acids respectively, and in adult fish, they were widely expressed in brain, pituitary, gonad, liver, kidney, head kidney, heart, muscle, gill, fin, eye and intestine. In male, both fsh and fshra transcripts showed high level of expression during spermatogenesis, however, in female, expression level was found to be higher in the fully grown oocyte stages. The expression of rohu lh and lhcgrba mRNA increased with increment of gonadosomatic index and showed highest level during spermiation stage in male and fully matured oocyte stage in female. These results together may suggest the involvement of fshra and lhcgrba in regulating function of seasonal gonadal development in rohu.
Collapse
MESH Headings
- Animals
- Cloning, Molecular
- Cyprinidae/genetics
- Cyprinidae/metabolism
- DNA, Complementary/isolation & purification
- DNA, Complementary/metabolism
- Female
- Gene Expression Profiling/veterinary
- Gonads/metabolism
- Male
- Pituitary Gland/metabolism
- Receptors, FSH/metabolism
- Receptors, Gonadotropin/genetics
- Receptors, Gonadotropin/isolation & purification
- Receptors, Gonadotropin/metabolism
- Receptors, LH/genetics
- Receptors, LH/metabolism
- Reproduction/genetics
- Sequence Analysis, DNA/veterinary
- Transcriptome
Collapse
Affiliation(s)
- Avinash Pradhan
- ICAR-Central Institute of Freshwater Aquaculture, Kausalyaganga, Bhubaneswar, Odisha, India
| | - Madhusmita Nayak
- ICAR-Central Institute of Freshwater Aquaculture, Kausalyaganga, Bhubaneswar, Odisha, India
| | - Mrinal Samanta
- ICAR-Central Institute of Freshwater Aquaculture, Kausalyaganga, Bhubaneswar, Odisha, India
| | - Rudra Prasanna Panda
- ICAR-Central Institute of Freshwater Aquaculture, Kausalyaganga, Bhubaneswar, Odisha, India
| | - Suresh Chandra Rath
- ICAR-Central Institute of Freshwater Aquaculture, Kausalyaganga, Bhubaneswar, Odisha, India
| | - Shiba Shankar Giri
- ICAR-Central Institute of Freshwater Aquaculture, Kausalyaganga, Bhubaneswar, Odisha, India
| | - Ashis Saha
- ICAR-Central Institute of Freshwater Aquaculture, Kausalyaganga, Bhubaneswar, Odisha, India.
| |
Collapse
|
40
|
Chen W, Liu L, Ge W. Expression analysis of growth differentiation factor 9 (Gdf9/gdf9), anti-müllerian hormone (Amh/amh) and aromatase (Cyp19a1a/cyp19a1a) during gonadal differentiation of the zebrafish, Danio rerio. Biol Reprod 2018; 96:401-413. [PMID: 28203731 DOI: 10.1095/biolreprod.116.144964] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 12/02/2016] [Accepted: 12/19/2016] [Indexed: 11/01/2022] Open
Abstract
In the zebrafish, no sex-determining gene has been identified, while some sex-related genes, such as cyp19a1a and amh, show sexually dimorphic expression. Interestingly, most of these genes are expressed in the somatic cells. With increasing evidence suggesting roles of germ cells in gonadal differentiation, there is an increasing interest in the factors released by the germ cells for the bidirectional communication between the two compartments. We have reported that Gdf9/gdf9 is an oocyte-specific factor in the zebrafish, similar to that of mammals. Whether and how Gdf9 is involved in gonadal differentiation is unknown. In this study, we compared the expression levels of gdf9, cyp19a1a, and amh among several other sex-related genes in the gonads before, during, and after sex differentiation. The expression of gdf9 started in the gonads before sex differentiation, and its level surged in the differentiated ovary. Its expression pattern was similar to that of cyp19a1a, but reciprocal to amh expression. Using recombinant zebrafish Gdf9 (rzfGdf9), we further showed that Gdf9 significantly suppressed the expression of amh while increased that of activin beta subunits (inhbaa and inhbb) in vitro. Although gdf9 and cyp19a1a showed co-expression during gonadal differentiation, we only observed a slight but not significant response of cyp19a1a to rzfGdf9. Knocking down the expression of gdf9 and cyp19a1a with vivo-morpholinos caused a male-skewed sex ratio. Our data suggested that Gdf9 is likely involved in promoting oocyte/ovary differentiation in the zebrafish and it may act by suppressing amh expression, at least partly, in the somatic cells.
Collapse
Affiliation(s)
- Weiting Chen
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau, China.,School of Life Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Lin Liu
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China.,School of Life Science, South China Normal University, Guangzhou, China
| | - Wei Ge
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau, China.,School of Life Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| |
Collapse
|
41
|
de Castro Assis LH, de Nóbrega RH, Gómez-González NE, Bogerd J, Schulz RW. Estrogen-induced inhibition of spermatogenesis in zebrafish is largely reversed by androgen. J Mol Endocrinol 2018; 60:273-284. [PMID: 29476039 DOI: 10.1530/jme-17-0177] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 02/22/2018] [Indexed: 12/20/2022]
Abstract
The hormonal regulation of spermatogenesis involves both gonadotropins and steroid hormones. Long-term in vivo exposure of adult zebrafish to estrogen impaired spermatogenesis associated with an androgen insufficiency, possibly induced by inhibiting gonadotropin release. Using this experimental model, we investigated if androgen treatment could enhance spermatogenesis, while maintaining the inhibition of gonadotropin release through continued estrogen exposure. Moreover, we also exposed animals to androgen alone, in order to examine androgen effects in the absence of estrogen-induced gonadotropin inhibition. Estrogen exposure depleted type B spermatogonia, meiotic and postmeiotic germ cells from the adult testis, but promoted the proliferation of type A undifferentiated spermatogonia, which accumulated in the testis. This change in germ cell composition was accompanied by reduced mRNA levels of those growth factors (e.g. insl3 and igf3) expressed by testicular somatic cells and known to stimulate spermatogonial differentiation in zebrafish. Additional androgen (11-ketoandrostenedione, which is converted to 11-ketotestosterone) treatment in vivo reversed most of the effects of estrogen exposure on spermatogenesis while insl3 and igf3 transcript levels remained suppressed. When androgen treatment was given alone, it promoted the production of haploid cells at the expense of spermatogonia, and increased transcript levels of some growth factor and hormone receptor genes, but not those of insl3 or igf3 We conclude that estrogen exposure efficiently inhibits spermatogenesis because it induces androgen insufficiency and suppresses gonadotropin-regulated growth factors known to stimulate germ cell differentiation. Moreover, our results suggest that androgens and the growth factors Insl3 and Igf3 stimulate spermatogenesis via independent pathways.
Collapse
Affiliation(s)
- Luiz Henrique de Castro Assis
- Reproductive Biology GroupDivision of Developmental Biology, Department of Biology, Faculty of Science, Institute of Biodynamics and Biocomplexity, Utrecht University, Utrecht, The Netherlands
| | - Rafael Henrique de Nóbrega
- Reproductive and Molecular Biology GroupDepartment of Morphology, Institute of Bioscience of Botucatu, São Paulo State University, Botucatu, São Paulo, Brazil
| | - Nuria Esther Gómez-González
- Department of Cell Biology and HistologyFaculty of Biology, University of Murcia, IMIB-Arrixaca, Murcia, Spain
| | - Jan Bogerd
- Reproductive Biology GroupDivision of Developmental Biology, Department of Biology, Faculty of Science, Institute of Biodynamics and Biocomplexity, Utrecht University, Utrecht, The Netherlands
| | - Rüdiger Winfried Schulz
- Reproductive Biology GroupDivision of Developmental Biology, Department of Biology, Faculty of Science, Institute of Biodynamics and Biocomplexity, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
42
|
Paredes JF, Lopez-Olmeda JF, Muñoz-Cueto JA, Sánchez-Vázquez F. Circadian expression of DNA methylation and demethylation genes in zebrafish gonads. Chronobiol Int 2018; 35:920-932. [PMID: 29509082 DOI: 10.1080/07420528.2018.1440403] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
This research aimed at investigating the light synchronization and endogenous origin of daily expression rhythms of eight key genes involved in epigenetic mechanisms (DNA methylation and demethylation) in zebrafish gonads. To this end, 84 zebrafish were distributed into six tanks, each one containing 14 fish (7 males and 7 females). Animals were subjected to 12 h light:12 h dark cycles (LD, lights on at ZT0 h) and fed randomly three times a day during the light phase. Locomotor activity rhythms were recorded in each tank for 20 days to test their synchronization to light. Then, zebrafish were fasted for one day and gonad samples were collected every 4 h during a 24 h cycle (ZT2, 6, 10, 14, 18, and 22 h). The results revealed that most of the epigenetic genes investigated exhibited a significant daily rhythm. DNA methylation genes (dnmt4, dnmt5, dnmt7) exhibited a daily rhythm of expression with a nocturnal acrophase (ZT14:01~ZT22:17 h), except for dnmt7 in males (ZT2:25 h). Similarly, all DNA demethylation genes (tet2, tdg, mb4, gadd45aa, and apobec2b) revealed the existence of statistically significant daily rhythms, except for gadd45aa in females. In females, tdg, mb4, and apobec2b presented a nocturnal peak (ZT14:20 ~ ZT22:04 h), whereas the tet2 acrophase was diurnal (ZT4:02 h). In males, tet2, tdg, and gadd45aa had nocturnal acrophases (ZT18:26~ZT21:31 h), whereas mb4 and apobec2b displayed diurnal acrophases (ZT5:28 and ZT4:02 h, respectively). To determine the endogenous nature of gene expression rhythms, another experiment was performed: 12 groups of 14 fish (7 males and 7 females) were kept in complete darkness (DD) and sampled every 4 h during a 48 h cycle (CT2, 6, 10, 14, 18, 22, 26, 30, 34, 38, 42, and 46 h). Under DD, most of the genes (7 out of 8) presented circadian rhythmicity with different endogenous periodicities (tau), suggesting that the epigenetic mechanisms of DNA methylation and demethylation in the gonads follow an internal control, functioning as part of the translation network linking the environment into somatic signals in fish reproduction.
Collapse
Affiliation(s)
| | - Jose Fernando Lopez-Olmeda
- b Department of Physiology, Faculty of Biology , University of Murcia , Murcia , Spain.,c Kalrsruhe Institute of Technology , Institute of Toxicology and Genetics , Eggenstein , Germany
| | - Jose A Muñoz-Cueto
- d Department of Biology, Faculty of Marine and Environmental Sciences, Faculty of Marine and Environmental Sciences , University of Cadiz , Cádiz , Spain.,e CACYTMAR , Research Institutes , Puerto Real , Spain
| | | |
Collapse
|
43
|
Li G, Tang H, Chen Y, Yin Y, Ogawa S, Liu M, Guo Y, Qi X, Liu Y, Parhar IS, Liu X, Lin H. Estrogen directly stimulates LHb expression at the pituitary level during puberty in female zebrafish. Mol Cell Endocrinol 2018; 461:1-11. [PMID: 28801227 DOI: 10.1016/j.mce.2017.08.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 08/05/2017] [Accepted: 08/06/2017] [Indexed: 11/16/2022]
Abstract
The LHb expression is up-regulated during puberty in female zebrafish. However, the molecular mechanism underlying how LHb expression is regulated during puberty remains largely unknown. In this study, we found that the mRNA expression levels of lhb, fshb and cyp19a1b were up-regulated along with the puberty onset in zebrafish. Among the three nuclear estrogen receptors (nERs), the esr2b is the only type whose expression is significantly up-regulated during puberty onset in the pituitary. However, in situ hybridization results revealed that lhb mRNA was colocalized with esr1 and esr2a but not esr2b. Exposure to estradiol (E2) significantly stimulates LHb expression in both wild-type and kiss1-/-;kiss2-/-;gnrh3-/- triple knockout pubertal zebrafish. Moreover, exposure of cultured pituitary cells to E2 increased the LHb expression, indicating that the estrogenic effect on LHb expression could be acted at the pituitary level. Finally, we cloned and analyzed the promoter of lhb by luciferase assay. Our results indicated that the E2 responsive regions of lhb promoter for ERα and ERβ2 are identical, suggesting that ERα and ERβ2 could bind to the same half ERE region of the promoter of lhb, exhibiting a classical ERE-dependent pathway. In summary, we demonstrate that E2 could directly act on the pituitary level to stimulate LHb transcription during puberty in zebrafish.
Collapse
Affiliation(s)
- Gaofei Li
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Haipei Tang
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Yu Chen
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Yike Yin
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Satoshi Ogawa
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Science, Monash University Malaysia, Bandar Sunway 47500, Malaysia
| | - Meifeng Liu
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Yin Guo
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Xin Qi
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Yun Liu
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Ishwar S Parhar
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Science, Monash University Malaysia, Bandar Sunway 47500, Malaysia
| | - Xiaochun Liu
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China; South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, China.
| | - Haoran Lin
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China; South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, China.
| |
Collapse
|
44
|
Zhu B, Ge W. Genome editing in fishes and their applications. Gen Comp Endocrinol 2018; 257:3-12. [PMID: 28919449 DOI: 10.1016/j.ygcen.2017.09.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Revised: 08/15/2017] [Accepted: 09/13/2017] [Indexed: 12/18/2022]
Abstract
There have been revolutionary progresses in genome engineering in the past few years. The newly-emerged genome editing technologies including zinc-finger nuclease (ZFN), transcription activator-like effector nuclease (TALEN) and clustered regularly interspaced short palindromic repeats associated with Cas9 (CRISPR/Cas9) have enabled biological scientists to perform efficient and precise targeted genome editing in different species. Fish represent the largest group of vertebrates with many species having values for both scientific research and aquaculture industry. Genome editing technologies have found extensive applications in different fish species for basic functional studies as well asapplied research in such fields as disease modeling and aquaculture. This mini-review focuses on recent advancements and applications of the new generation of genome editing technologies in fish species, with particular emphasis on their applications in understanding reproductive functions because the reproductive axis has been most systematically and best studied among others and its function has been difficult to address with reverse genetics approach.
Collapse
Affiliation(s)
- Bo Zhu
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Wei Ge
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau, China.
| |
Collapse
|
45
|
Using specific recombinant gonadotropins to induce spermatogenesis and spermiation in the European eel (Anguilla anguilla). Theriogenology 2018; 107:6-20. [DOI: 10.1016/j.theriogenology.2017.11.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 10/25/2017] [Accepted: 11/01/2017] [Indexed: 01/10/2023]
|
46
|
Zhu B, Pardeshi L, Chen Y, Ge W. Transcriptomic Analysis for Differentially Expressed Genes in Ovarian Follicle Activation in the Zebrafish. Front Endocrinol (Lausanne) 2018; 9:593. [PMID: 30364302 PMCID: PMC6193065 DOI: 10.3389/fendo.2018.00593] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 09/18/2018] [Indexed: 12/30/2022] Open
Abstract
In teleosts, the onset of puberty in females is marked by the appearance of the first wave of pre-vitellogenic (PV) follicles from the pool of primary growth (PG) follicles (follicle activation) in the ovary during sexual maturation. To understand the mechanisms underlying follicle activation and therefore puberty onset, we undertook this transcriptomic study to investigate gene expression profiles in the event. Our analysis revealed a total of 2,027 up-regulated and 859 down-regulated genes during the PG-PV transition. Gene Ontology (GO) analysis showed that in addition to basic cellular functions such as gene transcription, cell differentiation, and cell migration, other biological processes such as steroidogenesis, cell signaling and angiogenesis were also enriched in up-regulated genes; by comparison, some processes were down-regulated including piRNA metabolism, gene silencing and proteolysis. Further Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis identified a variety of signaling pathways that might play pivotal roles in PG-PV transition, including MAPK, TGF-β, Hedgehog, FoxO, VEGF, Jak-STAT, and phosphatidylinositol signaling pathways. Other pathways of particular interest included endocytosis and glycosaminoglycan biosynthesis. We also analyzed expression changes of genes expressed in different compartments viz. oocytes and follicle cells. Interestingly, most oocyte-specific genes remained unchanged in expression during follicle activation whereas a great number of genes specifically expressed in the follicle cells showed significant changes in expression. Overall, this study reported a comprehensive analysis for genes, biological processes and pathways involved in follicle activation, which also marks female puberty onset in the zebrafish when occurring for the first time in sexual maturation.
Collapse
Affiliation(s)
- Bo Zhu
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, China
| | - Lakhansing Pardeshi
- Genomics and Bioinformatics Core, Faculty of Health Sciences, University of Macau, Taipa, China
| | - Yingying Chen
- Genomics and Bioinformatics Core, Faculty of Health Sciences, University of Macau, Taipa, China
| | - Wei Ge
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, China
- *Correspondence: Wei Ge ;
| |
Collapse
|
47
|
Xie Y, Chu L, Liu Y, Sham KWY, Li J, Cheng CHK. The highly overlapping actions of Lh signaling and Fsh signaling on zebrafish spermatogenesis. J Endocrinol 2017; 234:233-246. [PMID: 28611209 DOI: 10.1530/joe-17-0079] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 06/09/2017] [Indexed: 01/30/2023]
Abstract
Gonadotropin signaling plays a pivotal role in the spermatogenesis of vertebrates, but exactly how gonadotropins regulate the process in non-mammalian species remains elusive. Using a gene knockout approach in zebrafish, we have previously demonstrated the non-canonical action of gonadotropin signaling on spermatogenesis by analyzing four single mutant lines (lhb, lhr, fshb and fshr) and three double mutant lines (lhb;fshb, lhr;fshr and fshb;lhr). In this study, we further investigated the actions of gonadotropins on the testis by establishing three other double-mutant zebrafish lines (lhb;lhr, fshb;fshr and lhb;fshr). All lhb;lhr and fshb;fshr mutant males were fertile. Analysis on the gonadosomatic index and testicular histology in these lhb;lhr and fshb;fshr mutants demonstrated that Lh signaling and Fsh signaling could functionally compensate each other in the testis. Intriguingly, it was found that the lhb;fshr mutant male fish were also morphologically and histologically normal and functionally fertile, a phenomenon which could be explained by the cross-activation of Lhr by Fsh. We have demonstrated this cross-reactivity for the first time in zebrafish. Fsh was shown to activate Lhr using three different assay systems, in which Lh-Fshr activation was also confirmed. Taken together, we conclude that the action of Lh signaling and Fsh signaling is redundant in that either alone can support zebrafish spermatogenesis based on two observations. First, that either Lh signaling or Fsh signaling alone is sufficient to support male fertility. Second, that the two gonadotropin ligands could promiscuously activate both receptors. Apart from revealing the complexity of gonadotropin signaling in controlling male reproduction in zebrafish, this study also shed light toward a better understanding on the evolution of gonadotropin signaling in vertebrates from fish to mammals.
Collapse
Affiliation(s)
- Yuxin Xie
- School of Biomedical SciencesThe Chinese University of Hong Kong-Shandong University Joint Laboratory on Reproductive Genetics, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - Lianhe Chu
- School of Biomedical SciencesThe Chinese University of Hong Kong-Shandong University Joint Laboratory on Reproductive Genetics, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - Yun Liu
- School of Biomedical SciencesThe Chinese University of Hong Kong-Shandong University Joint Laboratory on Reproductive Genetics, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - Kathy W Y Sham
- School of Biomedical SciencesThe Chinese University of Hong Kong-Shandong University Joint Laboratory on Reproductive Genetics, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - Jianzhen Li
- School of Biomedical SciencesThe Chinese University of Hong Kong-Shandong University Joint Laboratory on Reproductive Genetics, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
- College of Life SciencesNorthwest Normal University, Lanzhou, China
| | - Christopher H K Cheng
- School of Biomedical SciencesThe 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
|
48
|
Sevilhano T, de Carvalho RF, Oliveira NADJ, Oliveira JE, Maltarollo VG, Trossini G, Garcez R, Bartolini P. Molecular cloning and characterization of pirarucu (Arapaima gigas) follicle-stimulating hormone and luteinizing hormone β-subunit cDNAs. PLoS One 2017; 12:e0183545. [PMID: 28846736 PMCID: PMC5573580 DOI: 10.1371/journal.pone.0183545] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 08/07/2017] [Indexed: 02/04/2023] Open
Abstract
The common gonadotrophic hormone α-subunit (GTHα) has been previously isolated by our research group from A. gigas pituitaries; in the present work the cDNA sequences encoding FSHβ and LHβ subunits have also been isolated from the same species of fish. The FSH β-subunit consists of 126 amino acids with a putative 18 amino acid signal peptide and a 108 amino acid mature peptide, while the LH β-subunit consists of 141 amino acids with a putative 24 amino acid amino acid signal peptide and a 117 amino acid mature peptide. The highest identity, based on the amino acid sequences, was found with the order of Anguilliformes (61%) for FSHβ and of Cypriniformes (76%) for LHβ, followed by Siluriformes, 53% for FSHβ and 75% for LHβ. Interestingly, the identity with the corresponding human amino acid sequences was still remarkable: 45.1% for FSHβ and 51.4% for LHβ. Three dimensional models of ag-FSH and ag-LH, generated by using the crystal structures of h-FSH and h-LH as the respective templates and carried out via comparative modeling and molecular dynamics simulations, suggested the presence of the so-called "seat-belt", favored by a disulfide bond formed between the 3rd and 12th cysteine in both β-subunits. The sequences found will be used for the biotechnological synthesis of A. gigas gonadotrophic hormones (ag-FSH and ag-LH). In a first approach, to ascertain that the cloned transcripts allow the expression of the heterodimeric hormones, ag-FSH has been synthesized in human embryonic kidney 293 (HEK293) cells, preliminarily purified and characterized.
Collapse
Affiliation(s)
- Thais Sevilhano
- Biotechnology Department, IPEN-CNEN/SP, Cidade Universitária, São Paulo, SP, Brazil
| | | | | | | | | | - Gustavo Trossini
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Riviane Garcez
- Genetic Ichthyology Laboratory, Bioscience Institute, University of São Paulo, São Paulo, SP, Brazil
| | - Paolo Bartolini
- Biotechnology Department, IPEN-CNEN/SP, Cidade Universitária, São Paulo, SP, Brazil
| |
Collapse
|
49
|
de Jesus LWO, Bogerd J, Vieceli FM, Branco GS, Camargo MP, Cassel M, Moreira RG, Yan CYI, Borella MI. Gonadotropin subunits of the characiform Astyanax altiparanae: Molecular characterization, spatiotemporal expression and their possible role on female reproductive dysfunction in captivity. Gen Comp Endocrinol 2017; 246:150-163. [PMID: 27940043 DOI: 10.1016/j.ygcen.2016.12.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Revised: 12/02/2016] [Accepted: 12/06/2016] [Indexed: 10/20/2022]
Abstract
To better understand the endocrine control of reproduction in Characiformes and the reproductive dysfunctions that commonly occur in migratory fish of this order when kept in captivity, we chose Astyanax altiparanae, which has asynchronous ovarian development and multiple spawning events, as model species. From A. altiparanae pituitary total RNA, we cloned the full-length cDNAs coding for the follicle-stimulating hormone β subunit (fshb), the luteinizing hormone β subunit (lhb), and the common gonadotropin α subunit (gpha). All three sequences showed the highest degree of amino acid identity with other homologous sequences from Siluriformes and Cypriniformes. Real-time, quantitative PCR analysis showed that gpha, fshb and lhb mRNAs were restricted to the pituitary gland. In situ hybridization and immunofluorescence, using specific-developed and characterized polyclonal antibodies, revealed that both gonadotropin β subunits mRNAs/proteins are expressed by distinct populations of gonadotropic cells in the proximal pars distalis. No marked variations for lhb transcripts levels were detected during the reproductive cycle, and 17α,20β-dihydroxy-4-pregnen-3-one plasma levels were also constant, suggesting that the reproductive dysfunction seen in A. altiparanae females in captivity are probably due to a lack of increase of Lh synthesis during spawning season. In contrast, fshb transcripts changed significantly during the reproductive cycle, although estradiol-17β (E2) levels remained constant during the experiment, possibly due to a differential regulation of E2 synthesis. Taken together, these data demonstrate the putative involvement of gonadotropin signaling on the impairment of the reproductive function in a migratory species when kept in captivity. Future experimental studies must be carried to clarify this hypothesis. All these data open the possibility for further basic and applied studies related to reproduction in this fish model.
Collapse
Affiliation(s)
- Lázaro Wender O de Jesus
- Departamento de Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Professor Lineu Prestes 1524, 05508-000 São Paulo, SP, Brazil
| | - Jan Bogerd
- Reproductive Biology Group, Division Developmental Biology, Department of Biology, Faculty of Sciences, Utrecht University, Hugo R. Kruyt Building, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Felipe M Vieceli
- Departamento de Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Professor Lineu Prestes 1524, 05508-000 São Paulo, SP, Brazil
| | - Giovana S Branco
- Departamento de Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Professor Lineu Prestes 1524, 05508-000 São Paulo, SP, Brazil
| | - Marília P Camargo
- Departamento de Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Professor Lineu Prestes 1524, 05508-000 São Paulo, SP, Brazil
| | - Mônica Cassel
- Departamento de Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Professor Lineu Prestes 1524, 05508-000 São Paulo, SP, Brazil
| | - Renata G Moreira
- Departamento de Fisiologia, Instituto de Biociências, Universidade de São Paulo, Rua do Matão, Travessa 14, n.321, 05508-090 São Paulo, SP, Brazil
| | - Chao Y I Yan
- Departamento de Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Professor Lineu Prestes 1524, 05508-000 São Paulo, SP, Brazil
| | - Maria I Borella
- Departamento de Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Professor Lineu Prestes 1524, 05508-000 São Paulo, SP, Brazil.
| |
Collapse
|
50
|
Cowan M, Azpeleta C, López-Olmeda JF. Rhythms in the endocrine system of fish: a review. J Comp Physiol B 2017; 187:1057-1089. [DOI: 10.1007/s00360-017-1094-5] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Revised: 03/20/2017] [Accepted: 04/06/2017] [Indexed: 12/20/2022]
|