1
|
Guzmán A, Rosales-Torres AM, Medina-Moctezuma ZB, González-Aretia D, Hernández-Coronado CG. Effects and action mechanism of gonadotropins on ovarian follicular cells: A novel role of Sphingosine-1-Phosphate (S1P). A review. Gen Comp Endocrinol 2024; 357:114593. [PMID: 39047797 DOI: 10.1016/j.ygcen.2024.114593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 07/02/2024] [Accepted: 07/19/2024] [Indexed: 07/27/2024]
Abstract
Follicle-stimulating hormone (FSH) and luteinizing hormone (LH) control antral follicular growth by regulating several processes, such as the synthesis of hormones and signaling molecules, proliferation, survival, apoptosis, luteinization, and ovulation. To exert these effects, gonadotropins bind to their respective Gs protein-coupled receptors, activating the protein kinase A (PKA) pathway or recruiting Gq proteins to activate protein kinase C (PKC) signaling. Although the action mechanism of FSH and LH is clear, recently, it has been shown that both gonadotropins promote the synthesis of sphingosine-1-phosphate (S1P) in granulosa and theca cells through the activation of sphingosine kinase 1. Moreover, the inhibition of SPHKs reduces S1P synthesis, cell viability, and the proliferation of follicular cells in response to gonadotropins, and the addition of S1P to the culture medium increases the proliferation of granulosa and theca cells without apparent effects on sexual steroid synthesis. Therefore, we consider that S1P is a crucial signaling molecule that complements the canonical gonadotropin pathway to promote the proliferation and viability of granulosa and theca cells.
Collapse
Affiliation(s)
- A Guzmán
- Universidad Autónoma Metropolitana Unidad Xochimilco, Departamento Producción Agrícola y Animal, Ciudad de México, Mexico
| | - A M Rosales-Torres
- Universidad Autónoma Metropolitana Unidad Xochimilco, Departamento Producción Agrícola y Animal, Ciudad de México, Mexico
| | - Z B Medina-Moctezuma
- Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Ciudad de México, Mexico
| | - D González-Aretia
- Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Ciudad de México, Mexico
| | - C G Hernández-Coronado
- Universidad Autónoma Metropolitana Unidad Xochimilco, Departamento Producción Agrícola y Animal, Ciudad de México, Mexico.
| |
Collapse
|
2
|
Kodanch SM, Mukherjee S, Prabhu NB, Kabekkodu SP, Bhat SK, Rai PS. Altered mitochondrial homeostasis on bisphenol-A exposure and its association in developing polycystic ovary syndrome: A comprehensive review. Reprod Toxicol 2024; 130:108700. [PMID: 39181417 DOI: 10.1016/j.reprotox.2024.108700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Polycystic ovary syndrome (PCOS) is a heterogeneous endocrinopathy that is known to be one of the most common reproductive pathologies observed in premenopausal women around the globe and is particularly complex as it affects various endocrine and reproductive metabolic pathways. Endocrine-disrupting chemicals (EDCs) are considered to be environmental toxicants as they have hazardous health effects on the functioning of the human endocrine system. Among various classes of EDCs, bisphenol A (BPA) has been under meticulous investigation due to its ability to alter the endocrine processes. As there is emerging evidence suggesting that BPA-induced mitochondrial homeostasis dysfunction in various pathophysiological conditions, this review aims to provide a detailed review of how various pathways associated with ovarian mitochondrial homeostasis are impaired on BPA exposure and its mirroring effects on the PCOS phenotype. BPA exposure might cause significant damage to the mitochondrial morphology and functions through the generation of reactive oxygen species (ROS) and simultaneously downregulates the total antioxidant capacity, thereby leading to oxidative stress. BPA disrupts the mitochondrial dynamics in human cells by altering the expressions of mitochondrial fission and fusion genes, increases the senescence marker proteins, along with significant alterations in the mTOR/AMPK pathway, upregulates the expression of autophagy mediating factors, and downregulates the autophagic suppressor. Furthermore, an increase in apoptosis of the ovarian granulosa cells indicates impaired folliculogenesis. As all these key features are associated with the pathogenesis of PCOS, this review can provide a better insight into the possible associations between BPA-induced dysregulation of mitochondrial homeostasis and PCOS.
Collapse
Affiliation(s)
- Supraja M Kodanch
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Sayantani Mukherjee
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Navya B Prabhu
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Shashikala K Bhat
- Department of Obstetrics and Gynaecology, Dr T M A Pai Hospital, Udupi, Karnataka 576101, India
| | - Padmalatha S Rai
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India.
| |
Collapse
|
3
|
Xu W, Zou H, Zeng J, Mei W, Choi S. Effects of Various LED Light Spectra on Growth, Gonadal Development, and Growth-/Reproduction-Related Hormones in the Juvenile Red Spotted Grouper, Epinephelus akaara. Animals (Basel) 2023; 13:2047. [PMID: 37443845 DOI: 10.3390/ani13132047] [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: 05/11/2023] [Revised: 06/09/2023] [Accepted: 06/11/2023] [Indexed: 07/15/2023] Open
Abstract
The light spectrum is a key environmental cue involved in growth and reproduction in teleosts. This study investigated the effects of exposure on juvenile red spotted grouper exposed to white (control), red (590 nm), blue (480 nm), and green (520 nm) light-emitting diodes (LEDs) (12 h light:12 h dark) for two months. The body weight (BW), total length (TL), condition factor (CF), weight gain rate (WGR), gonadosomatic index (GSI), and hepatosomatic index (HSI) were assessed. Gonadal development was observed. The gene expression of growth-related hormones, such as growth hormone (GH), pre-pro-somatostatin-I (PSS-I), neuropeptide Y (NPY), and CCK, and of reproduction-related hormones, such as Kiss1, Kiss2, GPR54, sbGnRH, FSHβ, and LHβ, was analyzed. The results showed that the fish in the white LED group exhibited the best BW, TL, CF, WGR, and HSI after one or two months. The fish exposed to white LEDs showed the best growth after two months, but no significant differences in GH levels were detected. Contrarily, the expression levels of the PSS-I significantly increased (p < 0.05) in fish from the white group, suggesting the complex regulation of GH production and the limited effects of PSS-I on the inhibition of GH synthesis and somatic growth. The significantly increased NPY levels in the four LED groups (p < 0.05) indicated that these four LED spectra were effective in stimulating food intake and energy homeostasis. After two months, the gonads developed from chromatin nucleolar-stage oocytes to perinucleolar-stage oocytes in the four LED groups. The gene expression of Kiss2 and GPR54 in the four LED groups and of sbGnRH in the white and blue LED groups significantly increased when compared to that in the initial group (p < 0.05), while there were no significant differences in FSHβ and LHβ expression in the four LED groups. These results suggest that FSH and LH may not play important roles in gonadal development in juvenile red spotted grouper that are exposed to these four LED spectra.
Collapse
Affiliation(s)
- Wengang Xu
- School of Ocean, Yantai University, Yantai 264003, China
| | - Huafeng Zou
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China
| | - Jun Zeng
- Guangxi Academy of Sciences, Nanning 530007, China
- Institute of Beibu Gulf Marine Industry, Fangchenggang 538000, China
| | - Weiping Mei
- Guangxi Academy of Sciences, Nanning 530007, China
- Institute of Beibu Gulf Marine Industry, Fangchenggang 538000, China
| | - SongHee Choi
- Marine Science Institute, Jeju National University, Jeju 63333, Republic of Korea
| |
Collapse
|
4
|
Makowczenko KG, Jastrzebski JP, Kiezun M, Paukszto L, Dobrzyn K, Smolinska N, Kaminski T. Adaptation of the Porcine Pituitary Transcriptome, Spliceosome and Editome during Early Pregnancy. Int J Mol Sci 2023; 24:ijms24065946. [PMID: 36983019 PMCID: PMC10053595 DOI: 10.3390/ijms24065946] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/15/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
The physiological mechanisms of the porcine reproduction are relatively well-known. However, transcriptomic changes and the mechanisms accompanying transcription and translation processes in various reproductive organs, as well as their dependence on hormonal status, are still poorly understood. The aim of this study was to gain a principal understanding of alterations within the transcriptome, spliceosome and editome occurring in the pituitary of the domestic pig (Sus scrofa domestica L.), which controls basic physiological processes in the reproductive system. In this investigation, we performed extensive analyses of data obtained by high-throughput sequencing of RNA from the gilts' pituitary anterior lobes during embryo implantation and the mid-luteal phase of the estrous cycle. During analyses, we obtained detailed information on expression changes of 147 genes and 43 long noncoding RNAs, observed 784 alternative splicing events and also found the occurrence of 8729 allele-specific expression sites and 122 RNA editing events. The expression profiles of the selected 16 phenomena were confirmed by PCR or qPCR techniques. As a final result of functional meta-analysis, we acquired knowledge regarding intracellular pathways that induce changes in the processes accompanying transcription and translation regulation, which may induce modifications in the secretory activity of the porcine adenohypophyseal cells.
Collapse
Affiliation(s)
- Karol G Makowczenko
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Oczapowskiego 1A, 10-719 Olsztyn, Poland
| | - Jan P Jastrzebski
- Department of Plant Physiology, Genetics and Biotechnology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Oczapowskiego 1A, 10-719 Olsztyn, Poland
| | - Marta Kiezun
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Oczapowskiego 1A, 10-719 Olsztyn, Poland
| | - Lukasz Paukszto
- Department of Botany and Nature Protection, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Plac Lodzki 1, 10-719 Olsztyn, Poland
| | - Kamil Dobrzyn
- Department of Zoology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Oczapowskiego 5, 10-719 Olsztyn, Poland
| | - Nina Smolinska
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Oczapowskiego 1A, 10-719 Olsztyn, Poland
| | - Tadeusz Kaminski
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Oczapowskiego 1A, 10-719 Olsztyn, Poland
| |
Collapse
|
5
|
Wickramasuriya N, Hawkins R, Atwood C, Butler T. The roles of GnRH in the human central nervous system. Horm Behav 2022; 145:105230. [PMID: 35809386 PMCID: PMC9990468 DOI: 10.1016/j.yhbeh.2022.105230] [Citation(s) in RCA: 6] [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: 09/25/2021] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 11/04/2022]
Abstract
It is widely known that GnRH plays a role in facilitating reproductive function via the HPG axis, and this was once believed to be its only function. However, over the last several decades important neuromodulatory roles of GnRH in multiple brain functions have been elucidated. Multiple GnRH isoforms and receptors have been detected outside the HPG-axis across different species. In this review, we focus on the human CNS where GnRH I and II isoforms and a functional GnRH I receptor have been isolated. We first describe the traditional understanding of GnRH within the hypothalamus and the pituitary and current clinical use of GnRH analogues. We then review the location and function of GnRH-producing neurons and receptors located outside the HPG axis. We next review the GnRH I and II neuron location and quantity and GnRH I receptor gene expression throughout the human brain, using the Allen Brain Map Atlas. This analysis demonstrates a wide expression of GnRH throughout the brain, including prominent expression in the basal forebrain and cerebellum. Lastly, we examine the potential role of GnRH in aging and inflammation and its therapeutic potential for neurodegenerative disease and spinal cord lesions.
Collapse
Affiliation(s)
- Nimmi Wickramasuriya
- Weill Cornell Medicine, Department of Radiology, 1305 York Ave #3F, New York, NY 1002, USA
| | - Robert Hawkins
- Weill Cornell Medicine, Department of Radiology, 1305 York Ave #3F, New York, NY 1002, USA
| | - Craig Atwood
- University of Wisconsin, Department of Medicine, 2500 Overlook Tce, Madison, WI 53705, USA
| | - Tracy Butler
- Weill Cornell Medicine, Department of Radiology, 1305 York Ave #3F, New York, NY 1002, USA.
| |
Collapse
|
6
|
Batorek-Lukač N, Kress K, Čandek-Potokar M, Fazarinc G, Škrlep M, Poklukar K, Wesoly R, Stefanski V, Vrecl M. Immunocastration in adult boars as a model for late-onset hypogonadism. Andrology 2022; 10:1217-1232. [PMID: 35752946 PMCID: PMC9545940 DOI: 10.1111/andr.13219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 12/02/2022]
Abstract
Background While immunocastration has been studied in male pre‐pubertal pigs, data on older, sexually mature animals are limited. To understand the physiological effects of androgen deprivation in the late sexual development phase, we compared mature immunocastrated boars (n = 19; average age = 480 days) to young male immunocastrated pigs (n = 6; average age = 183 days) and young entire males (n = 6; average age = 186 days) as positive and negative controls, respectively. Objectives We hypothesized that the timing of gonadotropin‐releasing hormone suppression (early or late sexual development phases) influences the extent of reproductive function inhibition, histological structure of testicular tissue, and expression levels of selected genes related to steroid metabolism. Materials and methods Antibody titer, hormonal status, and histomorphometric analysis of testicular tissue were subjected to principal component analysis followed by hierarchical clustering to evaluate the immunocastration effectiveness in mature boars. Results Hierarchical clustering differentiated mature immunocastrated boars clustered with young immunocastrated pigs from those clustered with entire males. Although all mature immunocastrated boars responded to vaccination, as evidenced by the increased gonadotropin‐releasing hormone antibody titers (p < 0.001), decreased serum luteinizing hormone concentrations (p = 0.002), and changes in testicular tissue vascularization (lighter and less red testicular parenchyma; p ≤ 0.001), the responses were variable. Sharp decreases in testes index (p < 0.001), Leydig cell volume density (p < 0.001), Leydig cell nucleus‐to‐cytoplasm ratio (p < 0.001), and testosterone concentration (p < 0.001) were observed in mature immunocastrated boars clustered with young immunocastrated pigs compared with those that clustered with entire males. Additionally, mature immunocastrated boars clustered with young immunocastrated pigs showed lower hydroxysteroid 17‐beta dehydrogenase 7 expression than entire males (p < 0.05). The young immunocastrated pigs group showed higher follicle‐stimulating hormone receptors than the entire males and mature immunocastrated boars, lower steroidogenic acute regulatory protein expression levels compared with entire males, and mature immunocastrated boars clustered with entire males (p < 0.01). Conclusion The two‐dose vaccination regime resulted in progressive but variable regression of testicular function in adult (post‐pubertal) pigs; however, it was insufficient to induce a complete immunocastration response in all animals.
Collapse
Affiliation(s)
- Nina Batorek-Lukač
- Agricultural Institute of Slovenia, Hacquetova ulica 17, Ljubljana, SI-1000, Slovenia
| | - Kevin Kress
- University of Hohenheim, Garbenstraße 17, Stuttgart, 70599, Germany.,Schweinezuchtverband Baden-Württemberg e.V., Im Wolfer 10, Stuttgart, 70599, Germany
| | - Marjeta Čandek-Potokar
- Agricultural Institute of Slovenia, Hacquetova ulica 17, Ljubljana, SI-1000, Slovenia.,Faculty of Agriculture and Life Sciences, University of Maribor, Pivola 10, Hoče, SI-2311, Slovenia
| | - Gregor Fazarinc
- Veterinary Faculty, University of Ljubljana, Gerbičeva ulica 60, Ljubljana, SI-1000, Slovenia
| | - Martin Škrlep
- Agricultural Institute of Slovenia, Hacquetova ulica 17, Ljubljana, SI-1000, Slovenia
| | - Klavdija Poklukar
- Agricultural Institute of Slovenia, Hacquetova ulica 17, Ljubljana, SI-1000, Slovenia
| | - Raffael Wesoly
- Schweinezuchtverband Baden-Württemberg e.V., Im Wolfer 10, Stuttgart, 70599, Germany
| | - Volker Stefanski
- University of Hohenheim, Garbenstraße 17, Stuttgart, 70599, Germany
| | - Milka Vrecl
- Veterinary Faculty, University of Ljubljana, Gerbičeva ulica 60, Ljubljana, SI-1000, Slovenia
| |
Collapse
|
7
|
Jiang Y, Li S, Xu W, Ying J, Qu Y, Jiang X, Zhang A, Yue Y, Zhou R, Ruan T, Li J, Mu D. Critical Roles of the Circadian Transcription Factor BMAL1 in Reproductive Endocrinology and Fertility. Front Endocrinol (Lausanne) 2022; 13:818272. [PMID: 35311235 PMCID: PMC8924658 DOI: 10.3389/fendo.2022.818272] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 02/09/2022] [Indexed: 12/31/2022] Open
Abstract
Brain and muscle aryl-hydrocarbon receptor nuclear translocator like protein1 (BMAL1), a core component of circadian oscillation, is involved in many physiological activities. Increasing evidence has demonstrated the essential role of BMAL1 in reproductive physiology. For instance, BMAL1-knockout (KO) mice were infertile, with impaired reproductive organs and gametes. Additionally, in BMAL1-KO mice, hormone secretion and signaling of hypothalamus-pituitary-gonadal (H-P-G) hormones were also disrupted, indicating that H-P-G axis was impaired in BMAL1-KO mice. Moreover, both BMAL1-KO mice and BMAL1-knockdown by small interfering RNA (siRNA) in vitro cultured steroidogenic cells showed that BMAL1 was associated with gonadal steroidogenesis and expression of related genes. Importantly, BMAL1 also participates in pathogenesis of human reproductive diseases. In this review, we elaborate on the impaired reproduction of BMAL1-KO mice including the reproductive organs, reproductive endocrine hormones, and reproductive processes, highlighting the vital role of BMAL1 in fertility and reproductive endocrinology.
Collapse
Affiliation(s)
- Yin Jiang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Shiping Li
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Wenming Xu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
- Reproductive Endocrinology and Regulation Laboratory, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Junjie Ying
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Yi Qu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Xiaohui Jiang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
- Department of Andrology/Sichuan Human Sperm Bank, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Ayuan Zhang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Yan Yue
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Ruixi Zhou
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Tiechao Ruan
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Jinhui Li
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
- *Correspondence: Jinhui Li, ; Dezhi Mu,
| | - Dezhi Mu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
- *Correspondence: Jinhui Li, ; Dezhi Mu,
| |
Collapse
|
8
|
Tahir MS, Porto-Neto LR, Gondro C, Shittu OB, Wockner K, Tan AWL, Smith HR, Gouveia GC, Kour J, Fortes MRS. Meta-Analysis of Heifer Traits Identified Reproductive Pathways in Bos indicus Cattle. Genes (Basel) 2021; 12:768. [PMID: 34069992 PMCID: PMC8157873 DOI: 10.3390/genes12050768] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 12/12/2022] Open
Abstract
Fertility traits measured early in life define the reproductive potential of heifers. Knowledge of genetics and biology can help devise genomic selection methods to improve heifer fertility. In this study, we used ~2400 Brahman cattle to perform GWAS and multi-trait meta-analysis to determine genomic regions associated with heifer fertility. Heifer traits measured were pregnancy at first mating opportunity (PREG1, a binary trait), first conception score (FCS, score 1 to 3) and rebreeding score (REB, score 1 to 3.5). The heritability estimates were 0.17 (0.03) for PREG1, 0.11 (0.05) for FCS and 0.28 (0.05) for REB. The three traits were highly genetically correlated (0.75-0.83) as expected. Meta-analysis was performed using SNP effects estimated for each of the three traits, adjusted for standard error. We identified 1359 significant SNPs (p-value < 9.9 × 10-6 at FDR < 0.0001) in the multi-trait meta-analysis. Genomic regions of 0.5 Mb around each significant SNP from the meta-analysis were annotated to create a list of 2560 positional candidate genes. The most significant SNP was in the vicinity of a genomic region on chromosome 8, encompassing the genes SLC44A1, FSD1L, FKTN, TAL2 and TMEM38B. The genomic region in humans that contains homologs of these genes is associated with age at puberty in girls. Top significant SNPs pointed to additional fertility-related genes, again within a 0.5 Mb region, including ESR2, ITPR1, GNG2, RGS9BP, ANKRD27, TDRD12, GRM1, MTHFD1, PTGDR and NTNG1. Functional pathway enrichment analysis resulted in many positional candidate genes relating to known fertility pathways, including GnRH signaling, estrogen signaling, progesterone mediated oocyte maturation, cAMP signaling, calcium signaling, glutamatergic signaling, focal adhesion, PI3K-AKT signaling and ovarian steroidogenesis pathway. The comparison of results from this study with previous transcriptomics and proteomics studies on puberty of the same cattle breed (Brahman) but in a different population identified 392 genes in common from which some genes-BRAF, GABRA2, GABR1B, GAD1, FSHR, CNGA3, PDE10A, SNAP25, ESR2, GRIA2, ORAI1, EGFR, CHRNA5, VDAC2, ACVR2B, ORAI3, CYP11A1, GRIN2A, ATP2B3, CAMK2A, PLA2G, CAMK2D and MAPK3-are also part of the above-mentioned pathways. The biological functions of the positional candidate genes and their annotation to known pathways allowed integrating the results into a bigger picture of molecular mechanisms related to puberty in the hypothalamus-pituitary-ovarian axis. A reasonable number of genes, common between previous puberty studies and this study on early reproductive traits, corroborates the proposed molecular mechanisms. This study identified the polymorphism associated with early reproductive traits, and candidate genes that provided a visualization of the proposed mechanisms, coordinating the hypothalamic, pituitary, and ovarian functions for reproductive performance in Brahman cattle.
Collapse
Affiliation(s)
- Muhammad S. Tahir
- School of Chemistry and Molecular Bioscience, The University of Queensland Australia, Brisbane, QLD 4072, Australia; (M.S.T.); (O.B.S.); (K.W.); (A.W.L.T.); (H.R.S.); (J.K.)
| | - Laercio R. Porto-Neto
- Commonwealth Scientific and Industrial Research Organization, Brisbane, QLD 4072, Australia;
| | - Cedric Gondro
- Department of Animal Science, Michigan State University, East Lansing, MI 48824, USA;
| | - Olasege B. Shittu
- School of Chemistry and Molecular Bioscience, The University of Queensland Australia, Brisbane, QLD 4072, Australia; (M.S.T.); (O.B.S.); (K.W.); (A.W.L.T.); (H.R.S.); (J.K.)
| | - Kimberley Wockner
- School of Chemistry and Molecular Bioscience, The University of Queensland Australia, Brisbane, QLD 4072, Australia; (M.S.T.); (O.B.S.); (K.W.); (A.W.L.T.); (H.R.S.); (J.K.)
| | - Andre W. L. Tan
- School of Chemistry and Molecular Bioscience, The University of Queensland Australia, Brisbane, QLD 4072, Australia; (M.S.T.); (O.B.S.); (K.W.); (A.W.L.T.); (H.R.S.); (J.K.)
| | - Hugo R. Smith
- School of Chemistry and Molecular Bioscience, The University of Queensland Australia, Brisbane, QLD 4072, Australia; (M.S.T.); (O.B.S.); (K.W.); (A.W.L.T.); (H.R.S.); (J.K.)
| | - Gabriela C. Gouveia
- Animal Science Department, Veterinary School, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil;
| | - Jagish Kour
- School of Chemistry and Molecular Bioscience, The University of Queensland Australia, Brisbane, QLD 4072, Australia; (M.S.T.); (O.B.S.); (K.W.); (A.W.L.T.); (H.R.S.); (J.K.)
| | - Marina R. S. Fortes
- School of Chemistry and Molecular Bioscience, The University of Queensland Australia, Brisbane, QLD 4072, Australia; (M.S.T.); (O.B.S.); (K.W.); (A.W.L.T.); (H.R.S.); (J.K.)
| |
Collapse
|
9
|
Wang P, Sun Q, Wan R, Du Q, Xia X. Progesterone affects the transcription of genes in the circadian rhythm signaling and hypothalamic-pituitary-gonadal axes and changes the sex ratio in crucian carp (Carassius auratus). ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2020; 77:103378. [PMID: 32279014 DOI: 10.1016/j.etap.2020.103378] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 10/22/2019] [Accepted: 03/23/2020] [Indexed: 06/11/2023]
Abstract
Progesterone (P4) is an extensively applied progestin in human and veterinary medicine that has been widely detected in ambient aquatic environments, which can be detrimental to the health of aquatic organisms. Here we investigate the long-term effects of P4 on the transcription of genes related to the circadian rhythm signaling pathway and hypothalamic-pituitary-gonadal (HPG) axes in the crucian carp, which may have a potentially negative on endocrine-disrupting and sex differentiation impacts. Our results suggest that the expression of genes associated with the circadian rhythm signaling pathway are altered following exposure for 10, 20, 30, 40, 50 and 60 d, leading to disorders in the endocrine system disorders and the regulation of HPG axes-related gene expression. These maladies may affect gonadal development and the reproductive systems of crucian carp and provide a plausible mechanism for the observed change in sex ratio toward females after 180 d.
Collapse
Affiliation(s)
- Peijin Wang
- College of Life Science, Henan Normal University, Xinxiang, Henan 453007, People's Republic of China.
| | - Qingyu Sun
- College of Life Science, Henan Normal University, Xinxiang, Henan 453007, People's Republic of China.
| | - Ruyan Wan
- College of Life Science, Henan Normal University, Xinxiang, Henan 453007, People's Republic of China.
| | - Qiyan Du
- College of Life Science, Henan Normal University, Xinxiang, Henan 453007, People's Republic of China.
| | - Xiaohua Xia
- College of Life Science, Henan Normal University, Xinxiang, Henan 453007, People's Republic of China.
| |
Collapse
|
10
|
GnRH Antagonists Produce Differential Modulation of the Signaling Pathways Mediated by GnRH Receptors. Int J Mol Sci 2019; 20:ijms20225548. [PMID: 31703269 PMCID: PMC6888270 DOI: 10.3390/ijms20225548] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/29/2019] [Accepted: 11/04/2019] [Indexed: 12/13/2022] Open
Abstract
Commercial gonadotropin-releasing hormone (GnRH) antagonists differ by 1-2 amino acids and are used to inhibit gonadotropin production during assisted reproduction technologies (ART). In this study, potencies of three GnRH antagonists, Cetrorelix, Ganirelix and Teverelix, in inhibiting GnRH-mediated intracellular signaling, were compared in vitro. GnRH receptor (GnRHR)-transfected HEK293 and neuroblastoma-derived SH-SY5Y cell lines, as well as mouse pituitary LβT2 cells endogenously expressing the murine GnRHR, were treated with GnRH in the presence or absence of the antagonist. We evaluated intracellular calcium (Ca2+) and cAMP increases, cAMP-responsive element binding-protein (CREB) and extracellular-regulated kinase 1 and 2 (ERK1/2) phosphorylation, β-catenin activation and mouse luteinizing-hormone β-encoding gene (Lhb) transcription by bioluminescence resonance energy transfer (BRET), Western blotting, immunostaining and real-time PCR as appropriate. The kinetics of GnRH-induced Ca2+ rapid increase revealed dose-response accumulation with potency (EC50) of 23 nM in transfected HEK293 cells, transfected SH-SY5Y and LβT2 cells. Cetrorelix inhibited the 3 × EC50 GnRH-activated calcium signaling at concentrations of 1 nM-1 µM, demonstrating higher potency than Ganirelix and Teverelix, whose inhibitory doses fell within the 100 nM-1 µM range in both transfected HEK293 and SH-SY5Y cells in vitro. In transfected SH-SY5Y, Cetrorelix was also significantly more potent than other antagonists in reducing GnRH-mediated cAMP accumulation. All antagonists inhibited pERK1/2 and pCREB activation at similar doses, in LβT2 and transfected HEK293 cells treated with 100 nM GnRH. Although immunostainings suggested that Teverelix could be less effective than Cetrorelix and Ganirelix in inhibiting 1 µM GnRH-induced β-catenin activation, Lhb gene expression increase occurring upon LβT2 cell treatment by 1 µM GnRH was similarly inhibited by all antagonists. To conclude, this study has demonstrated Cetrorelix-, Ganirelix- and Teverelix-specific biased effects at the intracellular level, not affecting the efficacy of antagonists in inhibiting Lhb gene transcription.
Collapse
|
11
|
Xia X, Wang P, Wan R, Chang Z, Du Q. Progesterone affects sex differentiation and alters transcriptional of genes along circadian rhythm signaling and hypothalamic-pituitary-gonadal axes in juvenile Yellow River Carp (Cyprinus carpio var.). ENVIRONMENTAL TOXICOLOGY 2019; 34:1255-1262. [PMID: 31298479 DOI: 10.1002/tox.22826] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 06/14/2019] [Accepted: 06/26/2019] [Indexed: 06/10/2023]
Abstract
Progesterone (P4) is a biologically active steroid hormone that is involved in the regulation of oocyte growth and maturation, as well as development of the endometrium and implantation in the uterus of humans. It can also stimulate oocyte maturation in female fish, as well as spermatogenesis and sperm motility in male fish. Thus, P4 has been extensively used in human and animal husbandry as a typical progestin. However, P4 remaining in the water environment will pose a potential hazard to aquatic organisms. For example, it can interfere with sex differentiation and reproduction in aquatic vertebrates such as fish. Therefore, we investigated the effects of prolonged progesterone exposure on the expression of genes related to circadian rhythm signaling and the hypothalamic-pituitary-gonadal (HPG) axes in Yellow River Carp, which may have a potential impact on their sex differentiation. Our results suggested that P4 exposure altered the expression of genes related to circadian rhythm signaling, which can lead to disorders in the endocrine system and regulate the HPG axes-related activities. Furthermore, the expression of genes related to the HPG axes was also altered, which might affect gonadal development and the reproductive systems of Yellow River Carp. In addition, these changes may provide a plausible mechanism for the observed shifts in their sex ratio toward females.
Collapse
Affiliation(s)
- Xiaohua Xia
- College of Life Science, Henan Normal University, Xinxiang, China
| | - Peijin Wang
- College of Life Science, Henan Normal University, Xinxiang, China
| | - Ruyan Wan
- College of Life Science, Henan Normal University, Xinxiang, China
| | - Zhongjie Chang
- College of Life Science, Henan Normal University, Xinxiang, China
| | - Qiyan Du
- College of Life Science, Henan Normal University, Xinxiang, China
| |
Collapse
|
12
|
Zhang X, Yang W, Liang W, Wang Y, Zhang S. Intensity dependent disruptive effects of light at night on activation of the HPG axis of tree sparrows (Passer montanus). ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 249:904-909. [PMID: 30965542 DOI: 10.1016/j.envpol.2019.03.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 02/25/2019] [Accepted: 03/03/2019] [Indexed: 06/09/2023]
Abstract
Artificial light at night (ALAN) has become increasingly recognized as a disruptor of the reproductive endocrine process and behavior of wild birds. However, there is no evidence that ALAN directly disrupt the hypothalamus-pituitary-gonadal (HPG) axis, and no information on the effects of different ALAN intensities on birds. We experimentally tested whether ALAN affects reproductive endocrine activation in the HPG axis of birds, and whether this effect is related to the intensity of ALAN, in wild tree sparrows (Passer montanus). Forty-eight adult female birds were randomly assigned to four groups. They were first exposed to a short light photoperiod (8 h light and 16 h dark per day) for 20 days, then exposed to a long light photoperiod (16 h light and 8 h dark per day) to initiate the reproductive endocrine process. During these two kinds of photoperiod treatments, the four groups of birds were exposed to 0, 85, 150, and 300 lux light in the dark phase (night) respectively. The expression of the reproductive endocrine activation related TSH-β, Dio2 and GnRH-I gene was significantly higher in birds exposed to 85 lux light at night, and significantly lower in birds exposed to 150 and 300 lux, relative to the 0 lux control. The birds exposed to 85 lux had higher peak values of plasma LH and estradiol concentration and reached the peak earlier than birds exposed to 0, 150, or 300 lux did. The lower gene expression of birds exposed to 150 and 300 lux reduced their peak LH and estradiol values, but did not delay the timing of these peaks compared to the control group. These results reveal that low intensity ALAN accelerates the activation of the reproductive endocrine process in the HPG axis, whereas high intensity ALAN retards it.
Collapse
Affiliation(s)
- Xinjie Zhang
- College of Life and Environment Science, Minzu University of China, Beijing, 100081, China
| | - Wenyu Yang
- College of Life and Environment Science, Minzu University of China, Beijing, 100081, China
| | - Wei Liang
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, College of Life Sciences, Hainan Normal University, Haikou, 571158, China
| | - Yong Wang
- College of Agricultural, Life, and Natural Sciences, Alabama A&M University, Huntsville, AL, 35762, USA
| | - Shuping Zhang
- College of Life and Environment Science, Minzu University of China, Beijing, 100081, China.
| |
Collapse
|
13
|
Shi WJ, Hu LX, Huang GY, Liu YS, Zhang JN, Xie L, Ying GG. Dydrogesterone affects the transcription of genes in GnRH and steroidogenesis pathways and increases the frequency of atretic follicles in zebrafish (Danio rerio). CHEMOSPHERE 2019; 216:725-732. [PMID: 30391894 DOI: 10.1016/j.chemosphere.2018.10.202] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 10/28/2018] [Accepted: 10/29/2018] [Indexed: 06/08/2023]
Abstract
Dydrogesterone (DDG) is a synthetic progestin broadly used in human and veterinary medicine and has been widely detected in aquatic environments. However, its potential effects on aquatic organisms are little documented. Here we investigate the short-term effects of DDG on the transcriptional and histological responses in adult zebrafish (Danio rerio). Adult zebrafish were exposed to 32.0, 305 and 2490 ng L-1 of DDG for 14 days. Real time quantitative PCR analysis showed that DDG significantly increased transcripts of most genes involved in the gonadotropin-releasing hormone (GnRH) pathway in the brain of female. In contrast, apparent down-regulation of these gene transcriptions was observed in the brain of males. The transcription of cyp19a1a in the ovary had a 2.3 fold increase at 2490 ng L-1 of DDG and the transcription of hsd17b2 at 305 and 2490 ng L-1 in the testis was enhanced by approximately 2.0 fold and 2.4 fold, respectively. Histopathological analysis revealed exposure to 2490 ng L-1 DDG significantly increased the percentage of atretic follicles in the ovary. The results of this study suggest that DDG has potential endocrine disrupting effects and affects the ovarian development in zebrafish.
Collapse
Affiliation(s)
- Wen-Jun Shi
- The Environmental Research Institute, MOE Key Laboratory of Environmental Theoretical Chemistry, South China Normal University, Guangzhou 510006, China; State Key Laboratory of Organic Geochemistry, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China
| | - Li-Xin Hu
- The Environmental Research Institute, MOE Key Laboratory of Environmental Theoretical Chemistry, South China Normal University, Guangzhou 510006, China
| | - Guo-Yong Huang
- The Environmental Research Institute, MOE Key Laboratory of Environmental Theoretical Chemistry, South China Normal University, Guangzhou 510006, China
| | - You-Sheng Liu
- The Environmental Research Institute, MOE Key Laboratory of Environmental Theoretical Chemistry, South China Normal University, Guangzhou 510006, China
| | - Jin-Na Zhang
- State Key Laboratory of Organic Geochemistry, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China
| | - Lingtian Xie
- The Environmental Research Institute, MOE Key Laboratory of Environmental Theoretical Chemistry, South China Normal University, Guangzhou 510006, China
| | - Guang-Guo Ying
- The Environmental Research Institute, MOE Key Laboratory of Environmental Theoretical Chemistry, South China Normal University, Guangzhou 510006, China.
| |
Collapse
|
14
|
Chang JP, Pemberton JG. Comparative aspects of GnRH-Stimulated signal transduction in the vertebrate pituitary - Contributions from teleost model systems. Mol Cell Endocrinol 2018; 463:142-167. [PMID: 28587765 DOI: 10.1016/j.mce.2017.06.002] [Citation(s) in RCA: 37] [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: 03/20/2017] [Revised: 05/31/2017] [Accepted: 06/01/2017] [Indexed: 02/07/2023]
Abstract
Gonadotropin-releasing hormone (GnRH) is a major regulator of reproduction through actions on pituitary gonadotropin release and synthesis. Although it is often thought that pituitary cells are exposed to only one GnRH, multiple GnRH forms are delivered to the pituitary of teleost fishes; interestingly this can include the cGnRH-II form usually thought to be non-hypophysiotropic. GnRHs can regulate other pituitary cell-types, both directly as well as indirectly, and multiple GnRH receptors (GnRHRs) may also be expressed in the pituitary, and even within a single pituitary cell-type. Literature on the differential actions of native GnRH isoforms in primary pituitary cells is largely derived from teleost fishes. This review will outline the diversity and complexity of GnRH-GnRHR signal transduction found within vertebrate gonadotropes as well as extra-gonadotropic sites with special emphasis on comparative studies from fish models. The implications that GnRHR transduction mechanisms are GnRH isoform-, function-, and cell-specific are also discussed.
Collapse
Affiliation(s)
- John P Chang
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada.
| | - Joshua G Pemberton
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
15
|
Voliotis M, Garner KL, Alobaid H, Tsaneva-Atanasova K, McArdle CA. Gonadotropin-releasing hormone signaling: An information theoretic approach. Mol Cell Endocrinol 2018; 463:106-115. [PMID: 28760599 DOI: 10.1016/j.mce.2017.07.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 07/27/2017] [Accepted: 07/27/2017] [Indexed: 12/16/2022]
Abstract
Gonadotropin-releasing hormone (GnRH) is a peptide hormone that mediates central control of reproduction, acting via G-protein coupled receptors that are primarily Gq coupled and mediate GnRH effects on the synthesis and secretion of luteinizing hormone and follicle-stimulating hormone. A great deal is known about the GnRH receptor signaling network but GnRH is secreted in short pulses and much less is known about how gonadotropes decode this pulsatile signal. Similarly, single cell measures reveal considerable cell-cell heterogeneity in responses to GnRH but the impact of this variability on signaling is largely unknown. Ordinary differential equation-based mathematical models have been used to explore the decoding of pulse dynamics and information theory-derived statistical measures are increasingly used to address the influence of cell-cell variability on the amount of information transferred by signaling pathways. Here, we describe both approaches for GnRH signaling, with emphasis on novel insights gained from the information theoretic approach and on the fundamental question of why GnRH is secreted in pulses.
Collapse
Affiliation(s)
- Margaritis Voliotis
- Department of Mathematics and Living Systems Institute, College of Engineering, Mathematics and Physical Sciences, University of Exeter, Exeter, EX4 4QF, UK
| | - Kathryn L Garner
- Laboratories for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, University of Bristol, Whitson Street, Bristol, BS1 3NY, UK
| | - Hussah Alobaid
- Laboratories for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, University of Bristol, Whitson Street, Bristol, BS1 3NY, UK
| | - Krasimira Tsaneva-Atanasova
- Department of Mathematics and Living Systems Institute, College of Engineering, Mathematics and Physical Sciences, University of Exeter, Exeter, EX4 4QF, UK; EPSRC Centre for Predictive Modeling in Healthcare, University of Exeter, Exeter, EX4 4QF, UK
| | - Craig A McArdle
- Laboratories for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, University of Bristol, Whitson Street, Bristol, BS1 3NY, UK.
| |
Collapse
|
16
|
Shi WJ, Ying GG, Huang GY, Liang YQ, Hu LX, Zhao JL, Zhang JN. Transcriptional and Biochemical Alterations in Zebrafish Eleuthero-Embryos (Danio rerio) After Exposure to Synthetic Progestogen Dydrogesterone. BULLETIN OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2017; 99:39-45. [PMID: 28214940 DOI: 10.1007/s00128-017-2046-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 02/07/2017] [Indexed: 06/06/2023]
Abstract
Little information has so far been known on the effects of synthetic progestogen dydrogesterone (DDG) in organisms like fish. This study aimed to investigate the effects of DDG on the transcriptional and biochemical alterations in zebrafish eleuthero-embryos. Zebrafish eleuthero-embryos were analyzed for the transcriptional alterations by real-time quantitative PCR (RT-qPCR) and biochemical changes by attenuated total reflection Fourier-transform infrared spectroscopy (ATR-FITR) after 144 h exposure to DDG. The results of qPCR analysis showed that DDG exposure significantly suppressed the transcriptions of target genes involved in hypothalamic-pituitary-thyroid (HPT) axis, while it induced the expression of target genes mRNA belonging to hypothalamic-pituitary-gonad (HPG) axis. In addition, ATR-FTIR spectroscopy analysis showed that the biochemical alterations of protein, nucleic acid and lipid were observed following DDG treatment. The finding from this study suggests that DDG exposure could have potential multiple effects in fish.
Collapse
Affiliation(s)
- Wen-Jun Shi
- State Key Laboratory of Organic Geochemistry, CAS Research Centre of PRD Environmental Pollution and Control, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, 510640, Guangzhou, China
| | - Guang-Guo Ying
- State Key Laboratory of Organic Geochemistry, CAS Research Centre of PRD Environmental Pollution and Control, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, 510640, Guangzhou, China.
| | - Guo-Yong Huang
- State Key Laboratory of Organic Geochemistry, CAS Research Centre of PRD Environmental Pollution and Control, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, 510640, Guangzhou, China
| | - Yan-Qiu Liang
- State Key Laboratory of Organic Geochemistry, CAS Research Centre of PRD Environmental Pollution and Control, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, 510640, Guangzhou, China
- School of Chemistry and Environment, Guangdong Ocean University, 524088, Zhanjiang, China
| | - Li-Xin Hu
- State Key Laboratory of Organic Geochemistry, CAS Research Centre of PRD Environmental Pollution and Control, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, 510640, Guangzhou, China
| | - Jian-Liang Zhao
- State Key Laboratory of Organic Geochemistry, CAS Research Centre of PRD Environmental Pollution and Control, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, 510640, Guangzhou, China
| | - Jin-Na Zhang
- State Key Laboratory of Organic Geochemistry, CAS Research Centre of PRD Environmental Pollution and Control, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, 510640, Guangzhou, China
| |
Collapse
|
17
|
Desaulniers AT, Cederberg RA, Lents CA, White BR. Expression and Role of Gonadotropin-Releasing Hormone 2 and Its Receptor in Mammals. Front Endocrinol (Lausanne) 2017; 8:269. [PMID: 29312140 PMCID: PMC5732264 DOI: 10.3389/fendo.2017.00269] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 09/26/2017] [Indexed: 11/13/2022] Open
Abstract
Gonadotropin-releasing hormone 1 (GnRH1) and its receptor (GnRHR1) drive mammalian reproduction via regulation of the gonadotropins. Yet, a second form of GnRH (GnRH2) and its receptor (GnRHR2) also exist in mammals. GnRH2 has been completely conserved throughout 500 million years of evolution, signifying high selection pressure and a critical biological role. However, the GnRH2 gene is absent (e.g., rat) or inactivated (e.g., cow and sheep) in some species but retained in others (e.g., human, horse, and pig). Likewise, many species (e.g., human, chimpanzee, cow, and sheep) retain the GnRHR2 gene but lack the appropriate coding sequence to produce a full-length protein due to gene coding errors; although production of GnRHR2 in humans remains controversial. Certain mammals lack the GnRHR2 gene (e.g., mouse) or most exons entirely (e.g., rat). In contrast, old world monkeys, musk shrews, and pigs maintain the coding sequence required to produce a functional GnRHR2. Like GnRHR1, GnRHR2 is a 7-transmembrane, G protein-coupled receptor that interacts with Gαq/11 to mediate cell signaling. However, GnRHR2 retains a cytoplasmic tail and is only 40% homologous to GnRHR1. A role for GnRH2 and its receptor in mammals has been elusive, likely because common laboratory models lack both the ligand and receptor. Uniquely, both GnRH2 and GnRHR2 are ubiquitously expressed; transcript levels are abundant in peripheral tissues and scarcely found in regions of the brain associated with gonadotropin secretion, suggesting a divergent role from GnRH1/GnRHR1. Indeed, GnRH2 and its receptor are not physiological modulators of gonadotropin secretion in mammals. Instead, GnRH2 and GnRHR2 coordinate the interaction between nutritional status and sexual behavior in the female brain. Within peripheral tissues, GnRH2 and its receptor are novel regulators of reproductive organs. GnRH2 and GnRHR2 directly stimulate steroidogenesis within the porcine testis. In the female, GnRH2 and its receptor may help mediate placental function, implantation, and ovarian steroidogenesis. Furthermore, both the GnRH2 and GnRHR2 genes are expressed in human reproductive tumors and represent emerging targets for cancer treatment. Thus, GnRH2 and GnRHR2 have diverse functions in mammals which remain largely unexplored.
Collapse
Affiliation(s)
- Amy T. Desaulniers
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Rebecca A. Cederberg
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE, United States
| | | | - Brett R. White
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE, United States
- *Correspondence: Brett R. White,
| |
Collapse
|
18
|
Fernandez MO, Sharma S, Kim S, Rickert E, Hsueh K, Hwang V, Olefsky JM, Webster NJG. Obese Neuronal PPARγ Knockout Mice Are Leptin Sensitive but Show Impaired Glucose Tolerance and Fertility. Endocrinology 2017; 158:121-133. [PMID: 27841948 PMCID: PMC5412981 DOI: 10.1210/en.2016-1818] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 11/04/2016] [Indexed: 11/19/2022]
Abstract
The peroxisome-proliferator activated receptor γ (PPARγ) is expressed in the hypothalamus in areas involved in energy homeostasis and glucose metabolism. In this study, we created a deletion of PPARγ brain-knockout (BKO) in mature neurons in female mice to investigate its involvement in metabolism and reproduction. We observed that there was no difference in age at puberty onset between female BKOs and littermate controls, but the BKOs gave smaller litters when mated and fewer oocytes when ovulated. The female BKO mice had regular cycles but showed an increase in the number of cycles with prolonged estrus. The mice also had increased luteinizing hormone (LH) levels during the LH surge and histological examination showed hemorrhagic corpora lutea. The mice were challenged with a 60% high-fat diet (HFD). Metabolically, the female BKO mice showed normal body weight, glucose and insulin tolerance, and leptin levels but were protected from obesity-induced leptin resistance. The neuronal knockout also prevented the reduction in estrous cycles due to the HFD. Examination of ovarian histology showed a decrease in the number of primary and secondary follicles in both genotypes due to the HFD, but the BKO ovaries showed an increase in the number of hemorrhagic follicles. In summary, our results show that neuronal PPARγ is required for optimal female fertility but is also involved in the adverse effects of diet-induced obesity by creating leptin resistance potentially through induction of the repressor Socs3.
Collapse
Affiliation(s)
| | | | - Sun Kim
- Department of Medicine, School of Medicine, and
| | | | | | - Vicky Hwang
- Department of Medicine, School of Medicine, and
| | | | - Nicholas J G Webster
- Department of Medicine, School of Medicine, and
- Moores Cancer Center, University of California, San Diego, La Jolla, California 92093; and
- Medical Research Service, VA San Diego Healthcare System, San Diego, California 92161
| |
Collapse
|
19
|
Lin XW, Blum ID, Storch KF. Clocks within the Master Gland: Hypophyseal Rhythms and Their Physiological Significance. J Biol Rhythms 2015; 30:263-76. [PMID: 25926680 DOI: 10.1177/0748730415580881] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Various aspects of mammalian endocrine physiology show a time-of-day variation with a period of 24 h, which represents an adaptation to the daily environmental fluctuations resulting from the rotation of the earth. These 24-h rhythms in hormone abundance and consequently hormone function may rely on rhythmic signals produced by the master circadian clock, which resides in the suprachiasmatic nucleus and is thought to chiefly dictate the pattern of rest and activity in mammals in conjunction with the light/dark (LD) cycle. However, it is likely that clocks intrinsic to elements of the endocrine axes also contribute to the 24-h rhythms in hormone function. Here we review the evidence for rhythm generation in the endocrine master gland, the pituitary, and its physiological significance in the context of endocrine axes regulation and function.
Collapse
Affiliation(s)
- Xue-Wei Lin
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada Douglas Mental Health University Institute, Montreal, Quebec, Canada Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
| | - Ian David Blum
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada Douglas Mental Health University Institute, Montreal, Quebec, Canada Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
| | - Kai-Florian Storch
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada Douglas Mental Health University Institute, Montreal, Quebec, Canada
| |
Collapse
|
20
|
Millar RP, Babwah AV. KISS1R: Hallmarks of an Effective Regulator of the Neuroendocrine Axis. Neuroendocrinology 2015; 101:193-210. [PMID: 25765628 DOI: 10.1159/000381457] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 03/04/2015] [Indexed: 11/19/2022]
Abstract
Kisspeptin (KP) is now well recognized as a potent stimulator of gonadotropin-releasing hormone (GnRH) secretion and thereby a major regulator of the neuroendocrine-reproductive axis. KP signals via KISS1R, a G protein-coupled receptor (GPCR) that activates the G proteins Gαq/11. Modulation of the interaction of KP with KISS1R is therefore a potential new therapeutic target for stimulating (in infertility) or inhibiting (in hormone-dependent diseases) the reproductive hormone cascade. Major efforts are underway to target KISS1R in the treatment of sex steroid hormone-dependent disorders and to stimulate endogenous hormonal responses along the neuroendocrine axis as part of in vitro fertilization protocols. The development of analogs modulating KISS1R signaling will be aided by an understanding of the intracellular pathways and dynamics of KISS1R signaling under normal and pathological conditions. This review focuses on KISS1R recruitment of intracellular signaling (Gαq/11- and β-arrestin-dependent) pathways that mediate GnRH secretion and the respective roles of rapid desensitization, internalization, and recycling of resensitized receptors in maintaining an active population of KISS1R at the cell surface to facilitate prolonged KP signaling. Additionally, this review summarizes and discusses the major findings of an array of studies examining the desensitization of KP signaling in man, domestic and laboratory animals. This discussion highlights the major effects of ligand efficacy and concentration and the physiological, developmental, and metabolic status of the organism on KP signaling. Finally, the potential for the utilization of KP and analogs in stimulating and inhibiting the reproductive hormone cascade as an alternative to targeting the downstream GnRH receptor is discussed.
Collapse
Affiliation(s)
- Robert P Millar
- Mammal Research Institute, University of Pretoria, Pretoria, South Africa
| | | |
Collapse
|
21
|
Zhang S, Chen X, Zhang J, Li H. Differences in the reproductive hormone rhythm of tree sparrows (Passer montanus) from urban and rural sites in Beijing: the effect of anthropogenic light sources. Gen Comp Endocrinol 2014; 206:24-9. [PMID: 24861803 DOI: 10.1016/j.ygcen.2014.05.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 05/10/2014] [Accepted: 05/14/2014] [Indexed: 11/21/2022]
Abstract
The pervasiveness of anthropogenic light in urban environments has increased the exposure to light of many animals. Since photoperiod is a regulator of the timing of reproduction in most temperate region birds, such light sources could potentially change the timing of reproduction. We compared the luteinizing hormone (LH), testosterone (T), and estradiol (E2) levels of tree sparrow (Passer montanus) populations sampled at two urban and two rural sites in China, and also performed a controlled photoperiod experiment to determine the influence of artificial light on the endocrine rhythm of these populations. LH levels of urban tree sparrows increased earlier than those of rural ones, but rural populations had higher LH peaks. A linear mixed model (LMM) indicates that increased exposure to light at night (LAN) significantly influenced the LH, T and E2 concentrations of free-living tree sparrows in urban environments compared to their rural counterparts. The results of the controlled photoperiod experiment showed that tree sparrows that were exposed to 6lux of light during the dark phase of the artificial photoperiod began to secrete LH earlier, and had lower peak LH levels, than control birds. A LMM indicates that LAN had a significant effect on LH levels in this experiment. Although urban tree sparrows began to secrete LH earlier than their rural counterparts, we found no corresponding advance in T or E2 secretion. On the contrary, peak T and E2 levels of urban birds were lower than those of rural birds. These results suggest that although anthropogenic light sources appear to advance the onset of LH secretion in urban tree sparrow populations, they also lower peak LH, and consequently levels of T and E2. A possible explanation for these observations is that greater exposure to anthropogenic light in urban environments stimulates LH secretion and may influence photosensitivity, but further experimental work is required to test this hypothesis.
Collapse
Affiliation(s)
- Shuping Zhang
- College of Life and Environment Sciences, Minzu University of China, Beijing 100081, China.
| | - Xiaoyu Chen
- College of Life and Environment Sciences, Minzu University of China, Beijing 100081, China
| | - Jingruo Zhang
- College of Life and Environment Sciences, Minzu University of China, Beijing 100081, China
| | - Hongchang Li
- College of Life and Environment Sciences, Minzu University of China, Beijing 100081, China
| |
Collapse
|
22
|
Borromeo V, Berrini A, De Grandi F, Cremonesi F, Fiandanese N, Pocar P, Secchi C. A novel monoclonal antibody-based enzyme-linked immunosorbent assay to determine luteinizing hormone in bovine plasma. Domest Anim Endocrinol 2014; 48:145-57. [PMID: 24906940 DOI: 10.1016/j.domaniend.2014.03.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 03/25/2014] [Accepted: 03/30/2014] [Indexed: 10/25/2022]
Abstract
The development of a novel enzyme-linked immunosorbent assay (ELISA) for determining luteinizing hormone (LH) in bovine plasma is described. Anti-bovine LH (bLH) monoclonal antibodies (mAbs) were produced and characterized. One mAb recognizing the bLH β subunit was used for immunoaffinity purification of substantial amounts of biologically active bLH from pituitary glands. The purified bLH in combination with 2 anti-bLH β subunit mAbs was used to develop a sandwich ELISA, which satisfied all the criteria required to investigate LH secretory patterns in the bovine species. The ELISA standard curve was linear over the range 0.05 to 2.5 ng/mL, and the assay proved suitable for measuring bLH in plasma without any prior treatment of samples. Cross-reactivity and recovery tests confirmed the specificity of the method. The intra- and inter-assay coefficients of variation ranged between 3.41% and 9.40%, and 9.29% and 15.84%, respectively. The analytical specificity of the method was validated in vivo by provocative tests for LH in heifers, using the LH releasing peptide gonadotropin-releasing hormone. In conclusion, the adoption of mAbs for this ELISA for coating the wells and labeling, combined with the easy one-step production of reference bLH, ensures long-term continuity in large-scale measurements of LH in the bovine species.
Collapse
Affiliation(s)
- V Borromeo
- Dipartimento di Scienze Veterinarie e Sanità Pubblica, Università degli Studi, Milano, Italy.
| | - A Berrini
- Dipartimento di Scienze Veterinarie e Sanità Pubblica, Università degli Studi, Milano, Italy
| | - F De Grandi
- Dipartimento di Scienze Veterinarie e Sanità Pubblica, Università degli Studi, Milano, Italy
| | - F Cremonesi
- Dipartimento di Scienze Veterinarie per la Salute, la Produzione Animale e la Sicurezza Alimentare, Università degli Studi, Milano, Italy
| | - N Fiandanese
- Dipartimento di Scienze Veterinarie e Sanità Pubblica, Università degli Studi, Milano, Italy
| | - P Pocar
- Dipartimento di Scienze Veterinarie e Sanità Pubblica, Università degli Studi, Milano, Italy
| | - C Secchi
- Dipartimento di Scienze Veterinarie e Sanità Pubblica, Università degli Studi, Milano, Italy
| |
Collapse
|
23
|
Perrett RM, McArdle CA. Molecular mechanisms of gonadotropin-releasing hormone signaling: integrating cyclic nucleotides into the network. Front Endocrinol (Lausanne) 2013; 4:180. [PMID: 24312080 PMCID: PMC3834291 DOI: 10.3389/fendo.2013.00180] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 11/06/2013] [Indexed: 01/21/2023] Open
Abstract
Gonadotropin-releasing hormone (GnRH) is the primary regulator of mammalian reproductive function in both males and females. It acts via G-protein coupled receptors on gonadotropes to stimulate synthesis and secretion of the gonadotropin hormones luteinizing hormone and follicle-stimulating hormone. These receptors couple primarily via G-proteins of the Gq/ll family, driving activation of phospholipases C and mediating GnRH effects on gonadotropin synthesis and secretion. There is also good evidence that GnRH causes activation of other heterotrimeric G-proteins (Gs and Gi) with consequent effects on cyclic AMP production, as well as for effects on the soluble and particulate guanylyl cyclases that generate cGMP. Here we provide an overview of these pathways. We emphasize mechanisms underpinning pulsatile hormone signaling and the possible interplay of GnRH and autocrine or paracrine regulatory mechanisms in control of cyclic nucleotide signaling.
Collapse
Affiliation(s)
- Rebecca M. Perrett
- Laboratories for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, University of Bristol, Bristol, UK
| | - Craig A. McArdle
- Laboratories for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, University of Bristol, Bristol, UK
- *Correspondence: Craig A. McArdle, Laboratories for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, University of Bristol, 1 Whitson Street, Bristol BS1 3NY, UK e-mail:
| |
Collapse
|
24
|
Torrealday S, Lalioti MD, Guzeloglu-Kayisli O, Seli E. Characterization of the gonadotropin releasing hormone receptor (GnRHR) expression and activity in the female mouse ovary. Endocrinology 2013; 154:3877-87. [PMID: 23913446 PMCID: PMC3776864 DOI: 10.1210/en.2013-1341] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
GnRH agonists (GnRHa) are increasingly used for fertility preservation in women undergoing gonadotoxic chemotherapy. However, the protective mechanisms of action for these compounds have not yet been elucidated. In this study, we aimed to determine whether GnRHa have a direct effect on ovarian granulosa cells. GnRH receptor (GnRHR) expression was determined in mouse somatic and gonadal tissues including granulosa/cumulus cells and oocytes using quantitative RT-PCR and immunohistochemistry. Granulosa cells were isolated from mouse ovaries primed with pregnant mare serum gonadotropin. Response to GnRHa in cultured granulosa cells was assessed by determining the increase of intracellular cAMP and by assessing phosphorylation of downstream mediators of GnRH signaling: ERK and p38. To measure intracellular cAMP in our system, the cells were transfected with a cAMP-responsive luciferase reporter plasmid and stimulated with GnRHa. For all experiments, pituitary tissue and/or the αT3-1 mouse pituitary cell line were used as controls. GnRHR mRNA and protein were detected in mouse ovaries, granulosa/cumulus cells, and oocytes. After GnRHa stimulation at various time intervals, we were unable to detect a cAMP increase or activation of the ERK or p38 signaling pathway in cultured primary mouse granulosa cells, whereas activation was detected in the control αT3-1 mouse pituitary cells. In this study, we have not detected activation of the canonical GnRH signaling pathways in mouse ovarian somatic cells. Our findings suggest that the mechanism of action of GnRHa in the ovary is either below the detection level of our experimental design or is different from that in the pituitary.
Collapse
Affiliation(s)
- Saioa Torrealday
- MD, Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, 310 Cedar Street, LSOG 304B, New Haven, Connecticut 06520-8063.
| | | | | | | |
Collapse
|
25
|
Chu A, Zhu L, Blum ID, Mai O, Leliavski A, Fahrenkrug J, Oster H, Boehm U, Storch KF. Global but not gonadotrope-specific disruption of Bmal1 abolishes the luteinizing hormone surge without affecting ovulation. Endocrinology 2013; 154:2924-35. [PMID: 23736292 DOI: 10.1210/en.2013-1080] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Although there is evidence for a circadian regulation of the preovulatory LH surge, the contributions of individual tissue clocks to this process remain unclear. We studied female mice deficient in the Bmal1 gene (Bmal1(-/-)), which is essential for circadian clock function, and found that they lack the proestrous LH surge. However, spontaneous ovulation on the day of estrus was unaffected in these animals. Bmal1(-/-) females were also deficient in the proestrous FSH surge, which, like the LH surge, is GnRH-dependent. In the absence of circadian or external timing cues, Bmal1(-/-) females continued to cycle in constant darkness albeit with increased cycle length and time spent in estrus. Because pituitary gonadotropes are the source of circulating LH and FSH, we assessed hypophyseal circadian clock function and found that female pituitaries rhythmically express clock components throughout all cycle stages. To determine the role of the gonadotrope clock in the preovulatory LH and FSH surge process, we generated mice that specifically lack BMAL1 in gonadotropes (GBmal1KO). GBmal1KO females exhibited a modest elevation in both proestrous and baseline LH levels across all estrous stages. BMAL1 elimination from gonadotropes also led to increased variability in estrous cycle length, yet GBmal1KO animals were otherwise reproductively normal. Together our data suggest that the intrinsic clock in gonadotropes is dispensable for LH surge regulation but contributes to estrous cycle robustness. Thus, clocks in the suprachiasmatic nucleus or elsewhere must be involved in the generation of the LH surge, which, surprisingly, is not required for spontaneous ovulation.
Collapse
Affiliation(s)
- Adrienne Chu
- Douglas Mental Health University Institute and Department of Psychiatry, McGill University, Montreal, Quebec H4H 1R3, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Tang Y, He W, Wei Y, Qu Z, Zeng J, Qin C. Screening key genes and pathways in glioma based on gene set enrichment analysis and meta-analysis. J Mol Neurosci 2013; 50:324-32. [PMID: 23494636 DOI: 10.1007/s12031-013-9981-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 02/14/2013] [Indexed: 12/22/2022]
Abstract
Glioma is a highly invasive, rapidly spreading form of brain cancer, while its etiology is largely unknown. A few recently reported studies have been developed using gene expression microarrays of glioma to identify differentially expressed genes from several to hundreds. This study was designed to analyze vast amounts of glioma-related microarray data and screen the key genes and pathways related to the development and progression of glioma. We used gene set enrichment analysis (GSEA) and meta-analysis of seven included studies after standardized microarray preprocessing, which increased concordance between these gene datasets. After GSEA, there were 14 mixing pathways including 13 up- and 1 down-regulated pathways. Based on the meta-analysis, 268 significant genes were screened out (P < 0.05); there were 249 genes identified by Kyoto Encyclopedia of Genes and Genomes (KEGG), and 27 KEGG pathways closely related to the set of the imported genes were identified. At last, six consistent pathways and key genes in these pathways related to glioma were obtained with combined GSEA and meta-analysis. The gene pathways that we identified could provide insight concerning the development of glioma. Further studies are needed to determine the biological function for the positive genes.
Collapse
Affiliation(s)
- Yanyan Tang
- Department of Neurology, First Affiliated Hospital, Guangxi Medical University, No. 22, Shuang Yong Lu, Nanning 530021, Guangxi, China
| | | | | | | | | | | |
Collapse
|
27
|
A mathematical model of the bovine oestrous cycle: simulating outcomes of dietary and pharmacological interventions. J Theor Biol 2012; 313:115-26. [PMID: 22925571 DOI: 10.1016/j.jtbi.2012.08.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Revised: 05/16/2012] [Accepted: 08/08/2012] [Indexed: 11/20/2022]
Abstract
A mathematical model was constructed to simulate the bovine oestrous cycle by using nonlinear differential equations to describe the biological mechanisms which regulate the cycle. The model predicts circulating concentrations of gonadotrophin-releasing hormone, follicle-stimulating hormone, luteinizing hormone, oestradiol, inhibin and progesterone. These hormones collectively provide control and feedback mechanisms between the hypothalamus, pituitary gland and ovaries, which regulate ovarian follicular dynamics, corpus luteum function and ovulation. When follicular growth parameters are altered, the model predicts that cows will exhibit either two or three follicular waves per cycle, as seen in practice. Changes in other parameters allow the model to simulate: effects of nutrition on follicle recruitment and size of the ovulatory follicle; effects of negative energy balance on postpartum anoestrus; and effects of pharmacological intervention on hormone profiles and timing of ovulation. It is concluded that this model provides a sound basis for exploring factors that influence the bovine oestrous cycle in order to test hypotheses about nutritional and hormonal influences which, with further validation, should help to design dietary or pharmacological strategies for improving reproductive performance in cattle.
Collapse
|
28
|
Abstract
Fertility in dairy cows has been declining for the past three decades. Genetic selection for increased milk production has been associated with changes in key metabolic hormones (growth hormone, insulin, IGF and leptin) that regulate metabolism by homoeostasis and homeorhesis. These metabolic hormones, particularly insulin, provide signals to the reproductive system so that regulation of ovarian function is coordinated with changes in metabolic status. Studies have shown, for example, that increasing circulating insulin concentrations during the early post partum period can advance the resumption of oestrous cycles by enhancing follicular growth. However, high concentrations of insulin can be detrimental to the developmental competence of oocytes, which is also influenced by the supply of fatty acids at the systemic level and at the ovarian level. Insulin status is also associated with the incidence and characteristics of abnormal ovarian cycles. These changes can occur without significant variation in circulating gonadotrophin concentrations. This suggests that additional factors, such as peripheral metabolites, metabolic hormones and locally produced growth factors, may have a modulating role. Recent evidence has demonstrated that ovarian responses to metabolic signals and nutrient profile vary according to the stage of the reproductive cycle. Improved understanding of this multifactorial process enables nutrition to be matched to genotype and milk production, with a positive impact on pregnancy rate.
Collapse
|
29
|
Chen J, Zhang Y, Tang Z, Mao J, Kuang Z, Qin C, Li W. Production of recombinant orange-spotted grouper (Epinephelus coioides) follicle-stimulating hormone (FSH) in single-chain form and dimer form by Pichia pastoris and their biological activities. Gen Comp Endocrinol 2012; 178:237-49. [PMID: 22684083 DOI: 10.1016/j.ygcen.2012.05.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Revised: 05/24/2012] [Accepted: 05/29/2012] [Indexed: 11/21/2022]
Abstract
FSH is a key regulator of steroidogenesis and gonadal growth in teleosts. However, function of FSH is elusive in grouper due to the lack of purified and native FSH. In the present study, we reported production of bioactive orange-spotted grouper (Epinephelus coioides) FSH in dimer form and single-chain form by Pichia pastoris. Dimer form of recombinant grouper FSH (rgFSHba) was accomplished by co-expressing mature FSHb-subunit and a-subunit genes. Fusion of mature FSHb-subunit and a-subunit genes together linking with a polypeptide (4×(Gly-Ser)-Gly-Thr) gene generated single-chain form of recombinant grouper FSH (rgFSHb-a). Recombinant grouper common α-subunit (rgCga) and FSHb-subunit (rgFSHb) were also separately produced. Recombinant proteins were verified by Western blot and mass spectrometry assays, and characterized by deglycosylation analysis. Deglycosylation assay suggested that glycosylation of recombinant FSH mainly occurred on common a-subunit. Bioactivities of recombinant proteins were initially evaluated by activating grouper FSH receptor, and further demonstrated by incubating ovarian fragments of adult grouper and intraperitoneal injection in juvenile female grouper. Two forms of recombinant FSH presented similar biological activities of activating FSH receptor and stimulating in vitro testosterone (T) and estradiol-17β (E2) secretion, though the dimer form functioned slightly weaker than the single-chain form. However, injections of rgFSHb-a or rgFSHba could significantly increase serum T and E2 levels, induce early ovarian development, reduce hypothalamic gnrh1 mRNA level, and increase hypothalamic cyp19a1b mRNA level. Data in this study suggested that recombinant gonadotropin could be produced in dimer form or single-chain form by P. pastoris, and FSH could regulate steroidogenesis and early ovarian development in juvenile grouper.
Collapse
Affiliation(s)
- Jun Chen
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, Sun Yat-Sen University, Guangzhou 510275, China
| | | | | | | | | | | | | |
Collapse
|
30
|
Sirjani MA, Kohram H, Shahir MH. Effects of eCG injection combined with FSH and GnRH treatment on the lambing rate in synchronized Afshari ewes. Small Rumin Res 2012. [DOI: 10.1016/j.smallrumres.2012.04.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
31
|
Fatemi S, Mehrabani-Yeganeh H, Nejati-Javaremi A, Niknafs S. Association of neuropeptide Y and gonadotrophin-releasing hormone receptor gene SNPs with breeding value for growth and egg production traits in Mazandaran native chickens. GENETICS AND MOLECULAR RESEARCH 2012; 11:2539-47. [DOI: 10.4238/2012.july.10.9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
32
|
Jablonka-Shariff A, Boime I. A dileucine determinant in the carboxyl terminal sequence of the LHβ subunit is implicated in the regulated secretion of lutropin from transfected GH3 cells. Mol Cell Endocrinol 2011; 339:7-13. [PMID: 21458524 DOI: 10.1016/j.mce.2011.03.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2010] [Revised: 02/22/2011] [Accepted: 03/13/2011] [Indexed: 10/18/2022]
Abstract
LH and FSH are essential for control of gonadal function. They are synthesized in the same gonadotrope but differ in their mode of secretion. LH release is regulated, while FSH is secreted constitutively. One unique feature of LHβ is a carboxyl terminal hydrophobic heptapeptide. We demonstrated that deleting the heptapeptide diverted the truncated LH dimer to the constitutive pathway in vitro. To examine if the residues of this heptapeptide play a role in LH sorting, leucines 118-119 were substituted with alanine (L118A and L119A, respectively). The intracellular pool of the L118A mutant protein decreased with a corresponding increase in constitutive secretion. Moreover, immunofluorescence microscopy revealed that the L118A mutant exhibited fewer puncta as compared to wild-type LH. L119A behaved similar to wild-type LH, indicating that a single leucine residue at position 118, rather than a dileucine motif, contributes to the process that sorts LH into the regulated pathway.
Collapse
Affiliation(s)
- Albina Jablonka-Shariff
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | |
Collapse
|
33
|
Boer H, Stötzel C, Röblitz S, Deuflhard P, Veerkamp R, Woelders H. A simple mathematical model of the bovine estrous cycle: Follicle development and endocrine interactions. J Theor Biol 2011; 278:20-31. [DOI: 10.1016/j.jtbi.2011.02.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2010] [Revised: 12/22/2010] [Accepted: 02/23/2011] [Indexed: 10/18/2022]
|
34
|
Fernández M, Bourguignon N, Lux-Lantos V, Libertun C. Neonatal exposure to bisphenol a and reproductive and endocrine alterations resembling the polycystic ovarian syndrome in adult rats. ENVIRONMENTAL HEALTH PERSPECTIVES 2010; 118:1217-22. [PMID: 20413367 PMCID: PMC2944080 DOI: 10.1289/ehp.0901257] [Citation(s) in RCA: 167] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2009] [Accepted: 04/22/2010] [Indexed: 05/20/2023]
Abstract
BACKGROUND Bisphenol A (BPA), an endocrine disruptor, is a component of polycarbonate plastics, epoxy resins, and polystyrene. Several studies have reported potent in vivo effects, because BPA behaves as an estrogen agonist and/or antagonist and as an androgen and thyroid hormone antagonist. OBJECTIVES We investigated the effects of neonatal exposure to BPA on the reproductive axis in adult female Sprague-Dawley rats. METHODS Female rats were injected subcutaneously, daily from postnatal day 1 (PND1) to PND10 with BPA in castor oil at 500 microg/50 microL [BPA500; approximately 10-4 M, a dose higher than the lowest observed adverse effect level (LOAEL) of 50 mg/kg], 50 microg/50 microL (BPA50), or 5 microg/50 microL (both BPA50 and BPA5 are doses lower than the LOAEL), or castor oil vehicle alone. In adults we studied a) the release of gonadotropin-releasing hormone (GnRH) from hypothalamic explants, b) serum sex hormone levels, and c) ovarian morphology, ovulation, and fertility. RESULTS Neonatal exposure to BPA was associated with increased serum testosterone and estradiol levels, reduced progesterone in adulthood, and altered in vitro GnRH secretion. Animals exposed to BPA500 had altered ovarian morphology, showing a large number of cysts. Animals exposed to BPA50 had reduced fertility without changes in the number of oocytes on the morning of estrus, whereas animals exposed to BPA500 showed infertility. CONCLUSIONS Exposure to high doses of BPA during the period of brain sexual differentiation altered the hypothalamic-pituitary-gonadal axis in female Sprague-Dawley rats. These results have the potential to link neonatal exposure to high doses of BPA in rats with the development of polycystic ovarian syndrome. Studies of doses and routes of administration more consistent with human exposures are needed to determine the relevance of these findings to human health.
Collapse
Affiliation(s)
- Marina Fernández
- Instituto de Biología y Medicina Experimental, Buenos Aires, Argentina
- Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Nadia Bourguignon
- Instituto de Biología y Medicina Experimental, Buenos Aires, Argentina
| | | | - Carlos Libertun
- Instituto de Biología y Medicina Experimental, Buenos Aires, Argentina
- Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
- Address correspondence to C. Libertun, IByME-CONICET, Vuelta de Obligado 2490, (C1428ADN) Buenos Aires, Argentina. Telephone: 54-11-4783-2869. Fax: 54-11-4786-2564. E-mail:
| |
Collapse
|
35
|
Hirdes W, Dinu C, Bauer CK, Boehm U, Schwarz JR. Gonadotropin-releasing hormone inhibits ether-à-go-go-related gene K+ currents in mouse gonadotropes. Endocrinology 2010; 151:1079-88. [PMID: 20068004 DOI: 10.1210/en.2009-0718] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Secretion of LH from gonadotropes is initiated by a GnRH-induced increase in intracellular Ca(2+) concentration ([Ca(2+)](i)). This increase in [Ca(2+)](i) is the result of Ca(2+) release from intracellular stores and Ca(2+) influx through voltage-dependent Ca(2+) channels. Here we describe an ether-à-go-go-related gene (erg) K(+) current in primary mouse gonadotropes and its possible function in the control of Ca(2+) influx. To detect gonadotropes, we used a knock-in mouse strain, in which GnRH receptor-expressing cells are fluorescently labeled. Erg K(+) currents were recorded in 80-90% of gonadotropes. Blockage of erg currents by E-4031 depolarized the resting potential by 5-8 mV and led to an increase in [Ca(2+)](i), which was abolished by nifedipine. GnRH inhibited erg currents by a reduction of the maximal erg current and in some cells additionally by a shift of the activation curve to more positive potentials. In conclusion, the erg current contributes to the maintenance of the resting potential in gonadotropes, thereby securing a low [Ca(2+)](i) by restricting Ca(2+) influx. In addition, the erg channels are modulated by GnRH by an as-yet unknown signal cascade.
Collapse
Affiliation(s)
- Wiebke Hirdes
- Institute for Neural Signal Transduction, Center for Molecular Neurobiology, University Hospital Hamburg-Eppendorf, Falkenried 94, D-20253 Hamburg, Germany.
| | | | | | | | | |
Collapse
|
36
|
Pearl CA, Jablonka-Shariff A, Boime I. Rerouting of a follicle-stimulating hormone analog to the regulated secretory pathway. Endocrinology 2010; 151:388-93. [PMID: 19887562 PMCID: PMC2803143 DOI: 10.1210/en.2009-0939] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
LH and FSH are produced by the same gonadotrope cells of the anterior pituitary but differ in their mode of secretion. This coordinated secretion of LH and FSH is essential for normal follicular development and ovulation in females and for spermatogenesis in males. The structural signals encoded in the LH and FSH subunits that govern the intracellular sorting of LH through the regulated secretory pathway and FSH through the constitutive pathway are largely unknown. Our laboratory recently identified the seven amino acid carboxy tail of LH beta as a sorting signal for LH in GH(3) cells. Here we compared the morphological features of GH(3) cells expressing an FSH analog containing the heptapeptide (FL7AA) with wild-type FSH using confocal microscopy. These experiments were performed to develop a rerouting model for examining structure-function links between secretion pathways of FSH/LH and their biological action. Both FSH- and LH-expressing cells exhibit a fluorescence pattern of randomly dispersed cytoplasmic puncta. FL7AA expressing cells have more intracellular accumulation compared with wild-type FSH and display a unique halo pattern of fluorescence near the plasma membrane. Such a pattern was not observed in cells expressing FSH or LH. Our results demonstrate that this FSH analog containing the carboxy heptapeptide of LH beta is rerouted to the regulated secretory pathway in GH(3) cells. This rerouted gonadotropin provides a unique model to study the trafficking, regulation, and function of LH and FSH.
Collapse
Affiliation(s)
- Christopher A Pearl
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | |
Collapse
|
37
|
De Paul AL, Attademo AM, Carón RW, Soaje M, Torres AI, Jahn GA, Celis ME. Neuropeptide glutamic-isoleucine (NEI) specifically stimulates the secretory activity of gonadotrophs in primary cultures of female rat pituitary cells. Peptides 2009; 30:2081-7. [PMID: 19729046 DOI: 10.1016/j.peptides.2009.08.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2008] [Revised: 08/14/2009] [Accepted: 08/25/2009] [Indexed: 11/17/2022]
Abstract
The neuropeptide EI (NEI) is derived from proMCH. It activates GnRH neurons, and has been shown to stimulate the LH release following intracerebroventricular administration in several experimental models. The aim of the present paper was to evaluate NEI actions on pituitary hormone secretion and cell morphology in vitro. Pituitary cells from female rats were treated with NEI for a wide range of concentrations (1-400x10(-8)M) and time periods (1-5h). The media were collected and LH, FSH, PRL, and GH measured by RIA. The interaction between NEI (1, 10 and 100x10(-8)M) and GnRH (0.1 and 1x10(-9)M) was also tested. Pituitary cells were harvested for electron microscopy, and the immunogold immunocytochemistry of LH was assayed after 2 and 4h of NEI incubation. NEI (100x10(-8)M) induced a significant LH secretion after 2h of stimulus, reaching a maximum response 4h later. A rapid and remarkable LH release was induced by NEI (400x10(-8)M) 1h after stimulus, attaining its highest level at 2h. However, PRL, GH and FSH were not affected. NEI provoked ultrastructural changes in the gonadotrophs, which showed accumulations of LH-immunoreactive granules near the plasma membrane and exocytotic images, while the other populations exhibited no changes. Although NEI (10x10(-8)M), caused no action when used alone, its co-incubation with GnRH (1x10(-9)M), promoted a slight but significant increase in LH. These results demonstrate that NEI acts at the pituitary level through a direct action on gonadotrophs, as well as through interaction with GnRH.
Collapse
Affiliation(s)
- Ana Lucía De Paul
- Centro de Microscopía Electrónica, Universidad Nacional de Córdoba, Ciudad Universitaria, Argentina.
| | | | | | | | | | | | | |
Collapse
|
38
|
Pearl CA, Boime I. Sulfation of LH does not affect intracellular trafficking. Mol Cell Endocrinol 2009; 309:76-81. [PMID: 19647136 PMCID: PMC2720855 DOI: 10.1016/j.mce.2009.03.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2009] [Revised: 03/03/2009] [Accepted: 03/05/2009] [Indexed: 10/21/2022]
Abstract
LH and FSH are produced by the same gonadotrope cells of the anterior pituitary but differ in their mode of secretion. LH secretion is primarily episodic, or regulated, while FSH secretion is primarily basal, or constitutive. The asparagine (N)-linked oligosaccharides of LH and FSH terminate with sulfate and sialic acid, respectively. TSH also contains sulfated N-linked oligosaccharides and is secreted through the regulated pathway. It has been hypothesized that sulfate plays a role in segregating LH to the regulated pathway. Using a mouse pituitary model, we tested this hypothesis by examining the secretory fate of LH from pituitaries treated with sodium chlorate, a known inhibitor of sulfation. Here we show that mouse LH is sulfated and secreted through the regulated pathway, while FSH is secreted constitutively. LH secretion from chlorate-treated pituitaries, which showed complete inhibition of sulfation, was similar to untreated pituitaries. These data suggest that the metabolic role for sulfated N-linked oligosaccharides is not for intracellular trafficking but for the extracellular bioactivity of LH.
Collapse
Affiliation(s)
- Christopher A Pearl
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, United States
| | | |
Collapse
|
39
|
Matsuda M, Tsutsumi K, Kanematsu T, Fukami K, Terada Y, Takenawa T, Nakayama KI, Hirata M. Involvement of Phospholipase C-Related Inactive Protein in the Mouse Reproductive System Through the Regulation of Gonadotropin Levels1. Biol Reprod 2009; 81:681-9. [DOI: 10.1095/biolreprod.109.076760] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
40
|
Ramakrishnan S, Wayne NL. Social cues from conspecifics alter electrical activity of gonadotropin-releasing hormone neurons in the terminal nerve via visual signals. Am J Physiol Regul Integr Comp Physiol 2009; 297:R135-41. [DOI: 10.1152/ajpregu.00143.2009] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
There are multiple populations of gonadotropin-releasing hormone (GnRH) neurons in the brains of vertebrates. The population located in the hypothalamus/preoptic area is the best studied and is known to ultimately control reproduction. Teleost fish have an additional population of GnRH neurons in the terminal nerve (TN) associated with the olfactory bulbs, the physiological function of which is still unclear. Anatomical and physiological studies provide evidence that TN-GnRH neurons have extensive projections in the brain and modulate neuronal activity. Although there is anatomical evidence that the TN receives olfactory and optic sensory inputs, it is not known if sensory information is transmitted to TN-GnRH neurons to modulate their activity. In the present study, we tested the hypothesis that social cues from conspecifics modulate electrical activity of TN-GnRH neurons from the intact brain of female medaka fish ( Oryzias latipes). We further investigated the potential roles of chemosensory and visual signals in mediating the social cue response. We used a transgenic line of medaka with TN-GnRH neurons genetically tagged with green fluorescent protein, allowing visualization of specific neurons for whole-cell current clamp electrophysiology. We demonstrated that 24-h exposure to male visual and chemosensory cues suppressed the electrical activity of female TN-GnRH neurons compared with exposure to other females. Chemosensory cues alone were insufficient to induce this social cue response. However, visual cues alone replicated the “combined” social cue response. These findings support our hypothesis that sensory signals—and specifically, visual social cues—modulate electrical activity of TN-GnRH neurons.
Collapse
|
41
|
Fernández M, Bianchi M, Lux-Lantos V, Libertun C. Neonatal exposure to bisphenol a alters reproductive parameters and gonadotropin releasing hormone signaling in female rats. ENVIRONMENTAL HEALTH PERSPECTIVES 2009; 117:757-62. [PMID: 19479018 PMCID: PMC2685838 DOI: 10.1289/ehp.0800267] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2008] [Accepted: 01/06/2009] [Indexed: 05/03/2023]
Abstract
BACKGROUND Bisphenol A (BPA) is a component of polycarbonate plastics, epoxy resins, and polystyrene and is found in many products. Several reports have revealed potent in vivo effects, because BPA acts as an estrogen agonist and/or antagonist and as an androgen and thyroid hormone antagonist. OBJECTIVES We analyzed the effects of neonatal exposure to BPA on the reproductive axis of female Sprague-Dawley rats. METHODS Female rats were injected subcutaneously, daily, from postnatal day 1 (PND1) to PND10 with BPA [500 microg/50 microL (high) or 50 microg/50 microL (low)] in castor oil or with castor oil vehicle alone. We studied body weight and age at vaginal opening, estrous cycles, and pituitary hormone release in vivo and in vitro, as well as gonadotropin-releasing hormone (GnRH) pulsatility at PND13 and in adults. We also analyzed two GnRH-activated signaling pathways in the adults: inositol-triphosphate (IP(3)), and extracellular signal-regulated kinase(1/2) (ERK(1/2)). RESULTS Exposure to BPA altered pituitary function in infantile rats, lowering basal and GnRH-induced luteinizing hormone (LH) and increasing GnRH pulsatility. BPA dose-dependently accelerated puberty onset and altered estrous cyclicity, with the high dose causing permanent estrus. In adults treated neonatally with BPA, GnRH-induced LH secretion in vivo was decreased and GnRH pulsatility remained disrupted. In vitro, pituitary cells from animals treated with BPA showed lower basal LH and dose-dependently affected GnRH-induced IP(3) formation; the high dose also impaired GnRH-induced LH secretion. Both doses altered ERK(1/2) activation. CONCLUSIONS Neonatal exposure to BPA altered reproductive parameters and hypothalamic-pituitary function in female rats. To our knowledge, these results demonstrate for the first time that neonatal in vivo BPA permanently affects GnRH pulsatility and pituitary GnRH signaling.
Collapse
Affiliation(s)
- Marina Fernández
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Maria Bianchi
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Victoria Lux-Lantos
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Carlos Libertun
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Facultad de Medicina, Universidad de Buenos Aires, Argentina
- Address correspondence to C. Libertun, IByME-CONICET, Vuelta de Obligado 2490, (C1428ADN) Buenos Aires, Argentina. Telephone: 54-11-4783-2869. Fax: 54-11-4786-2564. E-mail:
| |
Collapse
|
42
|
Signaling by G-protein-coupled receptor (GPCR): studies on the GnRH receptor. Front Neuroendocrinol 2009; 30:10-29. [PMID: 18708085 DOI: 10.1016/j.yfrne.2008.07.001] [Citation(s) in RCA: 199] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2007] [Revised: 04/28/2008] [Accepted: 07/21/2008] [Indexed: 01/22/2023]
Abstract
Gonadotropin-releasing hormone (GnRH) is the first key hormone of reproduction. GnRH analogs are extensively used in in vitro fertilization, and treatment of sex hormone-dependent cancers, due to their ability to bring about 'chemical castration'. The interaction of GnRH with its cognate type I receptor (GnRHR) in pituitary gonadotropes results in the activation of Gq/G(11), phospholipase Cbeta (PLCbetaI), PLA(2), and PLD. Sequential activation of the phospholipases generates the second messengers inositol 1, 4, 5-trisphosphate (IP(3)), diacylglycerol (DAG), and arachidonic acid (AA), which are required for Ca(2+) mobilization, the activation of various protein kinase C isoforms (PKCs), and the production of prostaglandin (PG) and other metabolites of AA, respectively. PKC isoforms are the major mediators of the downstream activation of a number of mitogen-activated protein kinase (MAPK) cascades by GnRH, namely: extracellular signal-regulated kinase (ERK), jun-N-terminal kinase (JNK), and p38MAPK. The activated MAPKs phosphorylate both cytosolic and nuclear proteins to initiate the transcriptional activation of the gonadotropin subunit genes and the GnRHR. While Ca(2+) mobilization has been found to initiate rapid gonadotropin secretion, Ca(2+), together with various PKC isoforms, MAPKs and AA metabolites also serve as key nodes, in the GnRH-stimulated signaling network that enables the gonadotropes to decode GnRH pulse frequencies and translating that into differential gonadotropin synthesis and release. Even though pulsatility of GnRH is recognized as a major determinant for differential gonadotropin subunit gene expression and gonadotropin secretion very little is yet known about the signaling circuits governing GnRH action at the 'Systems Biology' level. Direct apoptotic and metastatic effects of GnRH analogs in gonadal steroid-dependent cancers expressing the GnRHR also seem to be mediated by the activation of the PKC/MAPK pathways. However, the mechanisms dictating life (pituitary) vs. death (cancer) decisions made by the same GnRHR remain elusive. Understanding these molecular mechanisms triggered by the GnRHR through biochemical and 'Systems Biology' approaches would provide the basis for the construction of the dynamic connectivity maps, which operate in the various cell types (endocrine, cancer, and immune system) targeted by GnRH. The connectivity maps will open a new vista for exploring the direct effects of GnRH analogs in tumors and the design of novel combined therapies for fertility control, reproductive disorders and cancers.
Collapse
|
43
|
Do MHT, Santos SJ, Lawson MA. GNRH induces the unfolded protein response in the LbetaT2 pituitary gonadotrope cell line. Mol Endocrinol 2008; 23:100-12. [PMID: 18974261 DOI: 10.1210/me.2008-0071] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The neuropeptide GNRH 1 stimulates the secretion of the reproductive hormone LH in pituitary gonadotropes. Other secretory cell types depend on the unfolded protein response (UPR) pathway to regulate protein synthesis and protect against endoplasmic reticulum (ER) stress in response to differentiation or secretory stimuli. This study investigated the role of the UPR in GNRH action within the LbetaT2 gonadotrope model. Cells were treated with GNRH, and the activation of UPR signaling components and general translational status was examined. The ER-resident stress sensors, Atf6, Eif2ak3, and Ern1, are all present, and GNRH stimulation results in the phosphorylation of eukaryotic translation initiation factor 2A kinase 3 and its downstream effector, eukaryotic translation initiation factor 2A. Additionally, activation of the UPR was confirmed both in LbetaT2 as well as mouse primary pituitary cells through identifying GNRH-induced splicing of Xbp1 mRNA, a transcription factor activated by splicing by the ER stress sensor, ER to nucleus signaling 1. Ribosome profiling revealed that GNRH stimulation caused a transient attenuation in translation, a hallmark of the UPR, remodeling ribosomes from actively translating polysomes to translationally inefficient ribonucleoprotein complexes and monosomes. The transient attenuation of specific mRNAs was also observed. Overall, the results show that GNRH activates components of the UPR pathway, and this pathway may play an important physiological role in adapting the ER of gonadotropes to the burden of their secretory demand.
Collapse
Affiliation(s)
- Minh-Ha T Do
- Department of Reproductive Medicine, Mail Code 0674, University of California, San Diego, La Jolla, California 92093-0674, USA
| | | | | |
Collapse
|
44
|
Martinez-Chavez CC, Minghetti M, Migaud H. GPR54 and rGnRH I gene expression during the onset of puberty in Nile tilapia. Gen Comp Endocrinol 2008; 156:224-33. [PMID: 18329643 DOI: 10.1016/j.ygcen.2008.01.019] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2007] [Revised: 01/28/2008] [Accepted: 01/31/2008] [Indexed: 11/16/2022]
Abstract
The Kiss1/GPR54 system has recently been shown to play a key role in the onset of puberty in mammals. Growing evidence suggests that this system is also conserved across vertebrates although very few studies so far have been performed in lower vertebrates. The aims of this study were firstly in the teleost Nile tilapia to screen tissues for GPR54 expression levels, secondly to measure the expression patterns of GPR54 and GnRH I receptor (rGnRH I) in whole brains during the onset of puberty and finally to determine the effects of continuous illumination (LL) on receptor expression levels. Results confirmed that GPR54 was predominantly expressed in the brain and pituitary of adult tilapia. Furthermore, a significant increase of GPR54 gene expression was found in tilapia brains at 11 weeks post hatch (wph) followed by rGnRH I at 13 wph just prior to the histological observation of vitellogenic oocytes and active spermatogenesis in ova and testes at 17 wph. These results suggest a correlation between the increase of GPR54 expression in the brain and the onset of puberty. Finally, a significant effect of LL was observed on GPR54 expression levels which were characterized by a delayed surge with significantly lower levels than those of control fish. The current study not only suggests a link between the Kiss1/GPR54 system and the onset of puberty in a tropical batch spawning teleost that would be a highly conserved feature across vertebrates but also that the transcriptional mechanisms regulating GPR54 expression could be directly or indirectly influenced by light.
Collapse
|
45
|
Robin E, Cognié J, Foulon-Gauze F, Fontaine J, Cayla X. Disruption of lipid rafts induces gonadotropin release in ovine pituitary and LbetaT2 gonadotroph cells. Biol Reprod 2008; 79:17-25. [PMID: 18322272 DOI: 10.1095/biolreprod.107.064881] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
In order to better understand the cellular mechanisms underlying LH and FSH secretion, we have addressed the contribution of lipid rafts to the secretion of gonadotropins. We used methyl-beta-cyclodextrin (MbetaCD), a cholesterol-sequestering agent, on an LbetaT2 murine gonadotroph cell line and on primary cultures of ovine pituitary cells. We found that in both systems, cholesterol depletion by MbetaCD induced a fast and substantial release of LH in the absence of natural stimulation by GnRH. In ovine pituitary cells, MbetaCD-mediated LH release was shown to be independent of protein synthesis. Twenty-four hours after MbetaCD treatment, there was no loss of cell viability and full recovery of LH secretory capabilities, as determined by GnRH or MbetaCD treatment. In addition, our data suggest the existence of a pool of LH that is not released by GnRH treatment but that is released by MbetaCD treatment. Finally, in ovine pituitary cells, MbetaCD treatment induced FSH secretion. Importantly, these in vitro data are supported by in vivo studies, because MbetaCD injected into the pituitary glands of anaesthetized sheep reproducibly induced a peak of LH release.
Collapse
Affiliation(s)
- E Robin
- UMR Physiologie de la Reproduction et des Comportements, INRA/CNRS/Université Tours/Haras Nationaux, 37380 Nouzilly, France
| | | | | | | | | |
Collapse
|
46
|
Pawson AJ, Faccenda E, Maudsley S, Lu ZL, Naor Z, Millar RP. Mammalian type I gonadotropin-releasing hormone receptors undergo slow, constitutive, agonist-independent internalization. Endocrinology 2008; 149:1415-22. [PMID: 18039780 DOI: 10.1210/en.2007-1159] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Regulatory elements present in the cytoplasmic carboxyl-terminal tails of G protein-coupled receptors contribute to agonist-dependent receptor desensitization, internalization, and association with accessory proteins such as beta-arrestin. The mammalian type I GnRH receptors are unique among the rhodopsin-like G protein-coupled receptors because they lack a cytoplasmic carboxyl-terminal tail. In addition, they do not recruit beta-arrestin, nor do they undergo rapid desensitization. By measuring the internalization of labeled GnRH agonists, previous studies have reported that mammalian type I GnRH receptors undergo slow agonist-dependent internalization. In the present study, we have measured the internalization of epitope-tagged GnRH receptors, both in the absence and presence of GnRH stimulation. We demonstrate that mammalian type I GnRH receptors exhibit a low level of constitutive agonist-independent internalization. Stimulation with GnRH agonist did not significantly enhance the level of receptor internalization above the constitutive level. In contrast, the catfish GnRH and rat TRH receptors, which have cytoplasmic carboxyl-terminal tails, displayed similar levels of constitutive agonist-independent internalization but underwent robust agonist-dependent internalization, as did chimeras of the mammalian type I GnRH receptor with the cytoplasmic carboxyl-terminal tails of the catfish GnRH receptor or the rat TRH receptor. When the carboxyl-terminal Tyr325 and Leu328 residues of the mammalian type I GnRH receptor were replaced with alanines, these two mutant receptors underwent significantly impaired internalization, suggesting a function for the Tyr-X-X-Leu sequence in mediating the constitutive agonist-independent internalization of mammalian type I GnRH receptors. These findings provide further support for the underlying notion that the absence of the cytoplasmic carboxyl-terminal tail of the mammalian type I GnRH receptors has been selected for during evolution to prevent rapid receptor desensitization and internalization to allow protracted GnRH signaling in mammals.
Collapse
Affiliation(s)
- Adam J Pawson
- Medical Research Council Human Reproductive Sciences Unit, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, United Kingdom.
| | | | | | | | | | | |
Collapse
|
47
|
Kitahara K, Sakai Y, Hosaka M, Hira Y, Kakizaki H, Watanabe T. Effects of a depot formulation of the GnRH agonist leuprorelin on the ultrastructure of male rat pituitary gonadotropes. ACTA ACUST UNITED AC 2007; 70:79-93. [PMID: 17827666 DOI: 10.1679/aohc.70.79] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
To clarify the acute and chronic effects of GnRH agonists on pituitary gonadotropes, changes in the ultrastructure of male rat gonadotropes were examined immunocytochemically and morphometrically after the administration of a one-month depot formulation of the GnRH agonist, leuprorelin. Immediately after the depot administration, the relative amounts of secretory granules drastically decreased in gonadotropes concomitantly with a marked increase in the plasma LH level. After the acute hyperstimulated phase, secretory granules in gonadotropes were gradually restored although the newly synthesized granules were less densely immunolabeled for LHbeta; their relative amounts and sizes were still significantly smaller than the controls after depot treatment for 28 days. Eighty-four days after the leuprorelin depot administration, however, the ultrastructural characteristics of pituitary gonadotropes appeared to recover as observed in controls: there were no significant differences in the relative amounts, sizes, and labeling densities for LHbeta of secretory granules, and the amounts of chromogranin A (CgA) and secretogranin II (SgII) were restored in secretory granules to control levels. When the rats were repeatedly treated with the leuprorelin depot at intervals of 4 weeks, the expression and intracellular storage levels of gonadotropins remained highly suppressed, judging from the labeling density for LHbeta. These findings suggest that the depot formulation of the GnRH agonist could suppress both the biosynthesis and release of gonadotropins for a month by synergistically depleting the intracellular storage of secretory granules at the onset of the treatment and by inducing the subsequent desensitization of the GnRH receptor signaling.
Collapse
|
48
|
Tannenbaum PL, Schultz-Darken NJ, Woller MJ, Abbott DH. Gonadotrophin-releasing hormone (GnRH) release in marmosets II: pulsatile release of GnRH and pituitary gonadotrophin in adult females. J Neuroendocrinol 2007; 19:354-63. [PMID: 17425610 DOI: 10.1111/j.1365-2826.2007.01535.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Unlike other mammals, including rodents, Old World primates and humans, common marmosets and probably all other New World primates synthesise and release chorionic gonadotrophin (CG), and not luteinising hormone (LH) from pituitary gonadotrophs. However, little is known about the physiological dynamics of gonadotrophin-releasing hormone (GnRH)-regulated CG release from gonadotrophs and whether such CG release has pulsatile release characteristics similar to those of LH in other mammalian species. Consequently, we performed a series of in vivo and in vitro studies in ovariectomised laboratory rats and female marmosets to compare GnRH-induced pituitary LH and CG release characteristics, respectively. Exogenous GnRH stimulated a slower onset of release of marmoset pituitary CG, both in vivo and in vitro, and induced an approximately 400% greater increase in the duration of marmoset pituitary CG release compared to that for rat LH. Not surprisingly, hypothalamic pulsatile release of GnRH in vivo was not obviously concordant with endogenous episodic changes in circulating levels of CG in marmosets, in contrast to the clear concordance observed between in vivo GnRH and LH release previously demonstrated in rats and other mammals. Pituitary CG release in marmosets thus demonstrates considerable divergence from the timely hypothalamic GnRH-regulated LH release in other female mammals, implying potentially different physiological dynamics in gonadotrophin regulation of marmoset ovarian function.
Collapse
Affiliation(s)
- P L Tannenbaum
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | | | | | | |
Collapse
|
49
|
Aizen J, Kasuto H, Golan M, Zakay H, Levavi-Sivan B. Tilapia Follicle-Stimulating Hormone (FSH): Immunochemistry, Stimulation by Gonadotropin-Releasing Hormone, and Effect of Biologically Active Recombinant FSH on Steroid Secretion1. Biol Reprod 2007; 76:692-700. [PMID: 17192515 DOI: 10.1095/biolreprod.106.055822] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
In fish, FSH is generally important for early gonadal development and vitellogenesis. As in mammals, FSH is a heterodimer composed of an alpha subunit that is noncovalently associated with the hormone-specific beta subunit. The objective of the present study was to express glycosylated, properly folded, and biologically active tilapia FSH (tFSH) using the Pichia pastoris expression system. Using this material, we aimed to develop a specific ELISA and to enable the study of FSH response to GnRH. The methylotrophic yeast P. pastoris was used to coexpress recombinant genes formed by fusion of mating factor alpha leader and tilapia fshb and cga coding sequences. Western blot analysis of tilapia pituitary FSH, resolved by SDS-PAGE, yielded a band of 15 kDa, while recombinant tFSH beta (rtFSH beta) and rtFSH beta alpha had molecular masses of 17-18 kDa and 26-30 kDa, respectively. Recombinant tFSH beta alpha was found to bear only N-linked carbohydrates. Recombinant tFSH beta alpha significantly enhanced 11-ketotestosterone (11-KT) and estradiol secretion from tilapia testes and ovaries, respectively, in a dose-dependent manner (similar to tilapia pituitary extract, affinity-purified pituitary FSH, and porcine FSH). Using antibodies raised against rtFSH beta, FSH-containing cells were localized adjacent to hypothalamic nerve fibers ramifying in the proximal pars distalis (PPD), while LH cells were localized in a more peripheral region of the PPD. Moreover, FSH is under the control of hypothalamic decapeptide GnRH, an effect that was abolished through the use of specific bioneutralizing antisera, anti-rtFSH beta. It also reduced basal secretion of 11-KT.
Collapse
Affiliation(s)
- Joseph Aizen
- Department of Animal Sciences, Faculty of Agricultural, Food and Environmental Quality Sciences, Hebrew University, Rehovot 76100, Israel
| | | | | | | | | |
Collapse
|
50
|
Abstract
Central roles in reproductive biology (i.e., growth and development of the oocyte, steroidogenesis, and ovulation) are played by the ovarian dominant follicle (DF). The DF is different from other follicles because it can escape atresia (the fate of all other follicles), and if exposed to the LH surge, its cells will differentiate into the corpus luteum. The DF was originally studied by looking at the surface of ovary through a surgical approach. Current studies employ a less-invasive ultrasound technique to track the growth and development of the DF. Recruitment and selection, the processes that give rise to the DF, and dominance, the physiological state of the mature DF, are important areas of basic research. Results of these basic studies are easily translated into real-world problems in farm animal reproduction. Superovulation, for example, overrides the selection mechanism and increases the number of ovulations. Understanding the factors that affect the size of the recruited pool should increase success rates (i.e., number of collected embryos) for superovulation. In most animals, the DF is short-lived, existing for long enough to allow for the final maturation of the oocyte. Some DF become atretic because they mature during the luteal phase and are never exposed to the LH surge. For other DF, the LH surge redirects the DF toward its ultimate demise (i.e., luteinization, ovulation, and differentiation into the corpus luteum). The DF is managed pharmacologically within protocols for timed AI. When timed AI fails, there may be abnormal corpus luteum development and early embryonic loss; outcomes that are secondary to inadequate follicular cell maturation and incomplete oocyte capacitation in the DF. Future work on the DF will clarify its underlying biological functions so that a variety of needs in farm animal reproduction can be efficiently managed.
Collapse
Affiliation(s)
- M C Lucy
- Department of Animal Sciences, University of Missouri, Columbia 65211
| |
Collapse
|