1
|
Barton BE, Erickson JA, Allred SI, Jeffries JM, Stephens KK, Hunter MI, Woodall KA, Winuthayanon W. Reversible female contraceptives: historical, current, and future perspectives†. Biol Reprod 2024; 110:14-32. [PMID: 37941453 PMCID: PMC10790348 DOI: 10.1093/biolre/ioad154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 10/30/2023] [Accepted: 11/01/2023] [Indexed: 11/10/2023] Open
Abstract
Contraception is a practice with extensive and complicated social and scientific histories. From cycle tracking, to the very first prescription contraceptive pill, to now having over-the-counter contraceptives on demand, family planning is an aspect of healthcare that has undergone and will continue to undergo several transformations through time. This review provides a comprehensive overview of current reversible hormonal and non-hormonal birth control methods as well as their mechanism of action, safety, and effectiveness specifically for individuals who can become pregnant. Additionally, we discuss the latest Food and Drug Administration (FDA)-approved hormonal method containing estetrol and drospirenone that has not yet been used worldwide as well as the first FDA-approved hormonal over-the-counter progestin-only pills. We also review available data on novel hormonal delivery through microchip, microneedle, and the latest FDA-approved non-hormonal methods such as vaginal pH regulators. Finally, this review will assist in advancing female contraceptive method development by underlining constructive directions for future pursuits. Information was gathered from the NCBI and Google Scholars databases using English and included publications from 1900 to present. Search terms included contraceptive names as well as efficacy, safety, and mechanism of action. In summary, we suggest that investigators consider the side effects and acceptability together with the efficacy of contraceptive candidate towards their development.
Collapse
Affiliation(s)
- Brooke E Barton
- School of Medicine, University of Washington, Seattle, WA, USA
| | - Jeffery A Erickson
- OB/GYN & Women’s Health, School of Medicine, University of Missouri, Columbia, MO, USA
- Translational Bioscience Program, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Stephanie I Allred
- OB/GYN & Women’s Health, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Jenna M Jeffries
- College of Art & Science, Washington State University, Pullman, WA, USA
| | - Kalli K Stephens
- OB/GYN & Women’s Health, School of Medicine, University of Missouri, Columbia, MO, USA
- Translational Bioscience Program, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Mark I Hunter
- OB/GYN & Women’s Health, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Kirby A Woodall
- OB/GYN & Women’s Health, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Wipawee Winuthayanon
- OB/GYN & Women’s Health, School of Medicine, University of Missouri, Columbia, MO, USA
| |
Collapse
|
2
|
Yang Y, Xu L, Lei B, Huang Y, Yu M. Effects of trichlorobisphenol A on the expression of proteins and genes associated with puberty initiation in GT1-7 cells and the relevant molecular mechanism. Food Chem Toxicol 2024; 183:114258. [PMID: 38040238 DOI: 10.1016/j.fct.2023.114258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/14/2023] [Accepted: 11/22/2023] [Indexed: 12/03/2023]
Abstract
This study evaluated the effects of Cl3BPA on kisspeptin-G-protein coupled receptor 54 (GPR54)/gonadotropin-releasing hormone (GnRH) (KGG) signals and analyzed the roles of estrogen receptor alpha (ERɑ) and G-protein coupled estrogen receptor 1 (GPER1) in regulating KGG signals. The results showed that Cl3BPA at 50 μM increased the levels of intracellular reactive oxygen species (ROS) and GnRH, upregulated the protein levels of kisspeptin and the expression of fshr, lhr and gnrh1 genes related to KGG in GT1-7 cells. In addition, 50 μM Cl3BPA significantly upregulated the phosphorylation of extracellular regulated protein kinases 1/2 (Erk1/2), the protein levels of GPER1 and the expression of the gper1 as well as the most target genes associated with mitogen-activated protein kinase (MAPK)/Erk1/2 pathways. Specific signal inhibitor experiments found that Cl3BPA activated KGG signals by activating the GPER1-mediated MAPK/Erk1/2 signaling pathway at the mRNA level. A docking test further confirmed the interactions between Cl3BPA and GPER1. The findings suggest that Cl3BPA might induce precocious puberty by increasing GnRH secretion together with KGG signaling upregulation, which is driven by GPER1-mediated signaling pathway. By comparison, ClxBPAs with fewer chlorine atoms had more obvious effects on the expression of proteins and partial genes related to KGG signals in GT1-7 cells.
Collapse
Affiliation(s)
- Yingxin Yang
- Institute of Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, PR China
| | - Lanbing Xu
- Institute of Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, PR China
| | - Bingli Lei
- Institute of Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, PR China.
| | - Yaoyao Huang
- Institute of Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, PR China
| | - Mengjie Yu
- Institute of Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, PR China
| |
Collapse
|
3
|
Functional Characterization of Three GnRH Isoforms in Small Yellow Croaker Larimichthys polyactis Maintained in Captivity: Special Emphasis on Reproductive Dysfunction. BIOLOGY 2022; 11:biology11081200. [PMID: 36009826 PMCID: PMC9404844 DOI: 10.3390/biology11081200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/07/2022] [Accepted: 08/09/2022] [Indexed: 11/16/2022]
Abstract
Fish reproduction is regulated by the brain–pituitary–gonad (BPG) axis where the gonadotropin-releasing hormone (GnRH) plays a central role. Seed production of small yellow croaker (Larimichthys polyactis) is performed using captive-reared broodstock known to undergo reproductive dysfunction, which is connected to endocrinological dysfunction. To determine the endocrinological mechanism of GnRHs in the BPG axis of small yellow croaker, full-length sequences of three GnRH isoforms encoding sbGnRH (GnRH1), cGnRH-II (GnRH2), and sGnRH (GnRH3) were cloned and characterized from brain tissue. qRT-PCR, in vivo, and in vitro experiments were performed for functional characterization. The mRNA expression of GnRH1 in the brain and gonadotropin subunits (GPα, FSHβ, and LHβ) in the pituitary were significantly higher at the ripen stage during gonadal development and GnRH1 at spawning stage during spawning events. Expression of both GnRH1 and GtH subunits was significantly lower in females than males. GtH subunits were induced at higher concentrations of GnRH1 in vivo and in vitro. Sex-steroids significantly inhibited the GnRH1 expression in vitro in a dose-dependent manner. Taken together, results indicated that GnRH1 plays a key role in gonadal maturation and sex-steroids induced negative feedback in the regulation of GnRH. A lower level of GnRH1 and GtHs might be responsible for reproductive dysfunction in a female small yellow croaker.
Collapse
|
4
|
Xu Y, Han CY, Park MJ, Gye MC. Increased testicular insulin-like growth factor 1 is associated with gonadal activation by recombinant growth hormone in immature rats. Reprod Biol Endocrinol 2022; 20:72. [PMID: 35459135 PMCID: PMC9034493 DOI: 10.1186/s12958-022-00944-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 04/11/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND In children, recombinant human growth hormone (rhGH) therapy for treatment of short stature has raised concerns of the early onset of puberty. Puberty is initiated by the activation of the hypothalamus-pituitary-gonad axis. Insulin-like growth factor-1 (IGF1) has been known to mediate physiologic effects of GH. To understand the mechanism of precocious sexual maturation following prepubertal GH therapy, the effects of rhGH on the hypothalamus-pituitary-gonad axis were examined in the immature male rats. METHODS Immature male rats were given by daily injection of rhGH (1 or 2 IU/kg) from postnatal day (PND) 21 to PND 23 or 30. The effects of rhGH on kisspeptin-GnRH-LH system in the hypothalamus-pituitary axis, systemic and testicular IGF1, spermatogenesis, steroidogenesis, and circulating testosterone levels were examined. The effects of rhGH on the IGF1 expression and steroidogenesis were examined in progenitor LCs in vitro. RESULTS Testicular steroidogenic pathway and spermatogenesis marker mRNA levels, number and size of 17β-hydroxysteroid dehydrogenase (+) LCs, and blood testosterone levels of rhGH rats were significantly higher than those of controls on PNDs 24 and 31. Hypothalamic Kiss1 and Gnrh1 mRNA of rhGH rats were significantly higher than those of controls on PND 24, indicating early activation of hypothalamic kisspeptin-GnRH neurons by rhGH. Hypothalamic Igf1 mRNA levels of rhGH rats were significantly higher than those of controls on PND 24 but significantly lower than those of controls on PND 31. Testicular Igf1 mRNA levels were significantly higher in rhGH rats than in the controls on PNDs 24 and 31 whereas circulating IGF1 levels were not. In progenitor LCs, rhGH significantly increased Igf1 and steroidogenic pathway mRNA levels and testosterone production. CONCLUSIONS Local increases in testicular IGF1 might be an important mediator of gonadal maturation via activation of LCs steroidogenesis in immature rats given rhGH.
Collapse
Affiliation(s)
- Yang Xu
- Department of Life Science and Institute for Natural Sciences, Hanyang University, Seoul, 04760, Korea
| | - Chang Yong Han
- Department of Life Science and Institute for Natural Sciences, Hanyang University, Seoul, 04760, Korea
| | - Mi Jung Park
- Department of Pediatrics, Sanggye Paik Hospital, Inje University College of Medicine, Seoul, 01757, Korea.
| | - Myung Chan Gye
- Department of Life Science and Institute for Natural Sciences, Hanyang University, Seoul, 04760, Korea.
| |
Collapse
|
5
|
Xiong J, Tian Y, Ling A, Liu Z, Zhao L, Cheng G. Genistein affects gonadotrophin-releasing hormone secretion in GT1-7 cells via modulating kisspeptin receptor and key regulators. Syst Biol Reprod Med 2022; 68:138-150. [PMID: 34986716 DOI: 10.1080/19396368.2021.2003910] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Epidemiological studies have shown that genistein, an isoflavonoid phytoestrogen from soybean, affects endocrine and reproductive systems and alters pubertal onset. Administration of genistein in mice could impact the electrophysiology of hypothalamic neurons associated with the secretion of gonadotropin-releasing hormone (GnRH), a key component of hypothalamic-pituitary-gonadal (HPG) axis that governs hormone release and reproductive maturation. However, whether genistein could directly influence GnRH secretion in GnRH-specific neurons requires further investigation. Here, mouse hypothalamic GT1-7 neurons were recruited as a GnRH-expressing model to directly evaluate the effect and mechanisms of genistein on GnRH release. Results from this study demonstrated that genistein treatment decreased cell viability, impacted cell cycle distribution, and induced apoptosis of GT1-7 cells. A high concentration of genistein (20 μM) significantly increased GnRH secretion by 122.4% compared to the control. Since GnRH release is regulated by components of the kisspeptin-neurokinin-dynorphin (KNDy) system and regulators including SIRT1, PKCγ, and MKRN3, their transcription and translation were examined. Significant increases were observed for the mRNA and protein levels of the KNDy component kisspeptin receptor (Gpr54/Kissr). Compared to the control, genistein treatment upregulated the level of Sirt1 mRNA level, while it downregulated Prkcg and Mkrn3 expression. Therefore, this study provided direct evidence that genistein treatment could affect GnRH secretion by modulating kisspeptin receptors, SIRT1, PKCγ and MKRN3 in GT1-7 cells.Abbreviations: GnRH: gonadotropin-releasing hormone; HPG: hypothalamic-pituitary-gonadal; KNDy: kisspeptin-neurokinin-dynorphin; LH: luteinizing hormone; FSH: follicle-stimulating hormone; ARC: arcuate nucleus; ER: estrogen receptor; SIRT1: silent information regulator 1; PKCγ: protein kinase c γ: MKRN3: makorin ring finger protein 3; LC: lethal concentration; PI: propidium iodide; ECL: chemiluminescence; BCA: bicinchoninic acid assay; PBS: phosphate-buffered saline; CT: fluorescence reached threshold; PVDF: polyvinylidene difluoride.
Collapse
Affiliation(s)
- Jingyuan Xiong
- Healthy Food Evaluation Research Center, Department of Occupational and Environmental Health, Department of Maternal, Child and Adolescent Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Ye Tian
- Healthy Food Evaluation Research Center, Department of Occupational and Environmental Health, Department of Maternal, Child and Adolescent Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Aru Ling
- Laboratory of Molecular Translational Medicine, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Zhenmi Liu
- Healthy Food Evaluation Research Center, Department of Occupational and Environmental Health, Department of Maternal, Child and Adolescent Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Li Zhao
- Healthy Food Evaluation Research Center, Department of Occupational and Environmental Health, Department of Maternal, Child and Adolescent Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Guo Cheng
- Laboratory of Molecular Translational Medicine, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
6
|
Lopez-Rodriguez D, Franssen D, Bakker J, Lomniczi A, Parent AS. Cellular and molecular features of EDC exposure: consequences for the GnRH network. Nat Rev Endocrinol 2021; 17:83-96. [PMID: 33288917 DOI: 10.1038/s41574-020-00436-3] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/16/2020] [Indexed: 12/12/2022]
Abstract
The onset of puberty and the female ovulatory cycle are important developmental milestones of the reproductive system. These processes are controlled by a tightly organized network of neurotransmitters and neuropeptides, as well as genetic, epigenetic and hormonal factors, which ultimately drive the pulsatile secretion of gonadotropin-releasing hormone. They also strongly depend on organizational processes that take place during fetal and early postnatal life. Therefore, exposure to environmental pollutants such as endocrine-disrupting chemicals (EDCs) during critical periods of development can result in altered brain development, delayed or advanced puberty and long-term reproductive consequences, such as impaired fertility. The gonads and peripheral organs are targets of EDCs, and research from the past few years suggests that the organization of the neuroendocrine control of reproduction is also sensitive to environmental cues and disruption. Among other mechanisms, EDCs interfere with the action of steroidal and non-steroidal receptors, and alter enzymatic, metabolic and epigenetic pathways during development. In this Review, we discuss the cellular and molecular consequences of perinatal exposure (mostly in rodents) to representative EDCs with a focus on the neuroendocrine control of reproduction, pubertal timing and the female ovulatory cycle.
Collapse
Affiliation(s)
| | - Delphine Franssen
- Neuroendocrinology Unit, GIGA Neurosciences, University of Liège, Liège, Belgium
| | - Julie Bakker
- Neuroendocrinology Unit, GIGA Neurosciences, University of Liège, Liège, Belgium
| | - Alejandro Lomniczi
- Division of Neuroscience, Oregon National Primate Research Center (ONPRC), OHSU, OR, USA
| | - Anne-Simone Parent
- Neuroendocrinology Unit, GIGA Neurosciences, University of Liège, Liège, Belgium.
- Department of Pediatrics, University Hospital Liège, Liège, Belgium.
| |
Collapse
|
7
|
Chen HP, Cui XF, Wang YR, Li ZY, Tian CX, Jiang DN, Zhu CH, Zhang Y, Li SS, Li GL. Identification, functional characterization, and estrogen regulation on gonadotropin-releasing hormone in the spotted scat, Scatophagus argus. FISH PHYSIOLOGY AND BIOCHEMISTRY 2020; 46:1743-1757. [PMID: 32514853 DOI: 10.1007/s10695-020-00825-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 05/20/2020] [Indexed: 06/11/2023]
Abstract
Gonadotropin-releasing hormone (GnRH) is a key neuropeptide of the reproductive system. However, little is known about the role of GnRH in the spotted scat (Scatophagus argus). Here, three GnRH subtypes (cGnRH-II, sGnRH, and sbGnRH) were identified in the spotted scat. cGnRH-II and sGnRH were only expressed in the brains and gonads of both male and female fish, exhibiting a tissue-specific expression pattern, while sbGnRH was expressed at different transcription levels in all examined tissues. During ovarian maturation, hypothalamus-associated sbGnRH was upregulated, while the expression of sGnRH was variable and cGnRH-II first increased and then decreased. In vivo experiments showed that sbGnRH significantly promoted the expression of fsh and lh genes in a dose-dependent manner and exhibited a desensitization effect on lh expression at high concentrations. For sGnRH and cGnRH-II, only high concentrations could induce fsh and lh expression. Furthermore, treatment with highly concentrated sbGnRH peptide also induced fsh and lh expression, whereas the sGnRH and cGnRH-II peptides only induced fsh expression in vitro. 17β-Estradiol (E2) significantly inhibited the expression of sbGnRH mRNA in a dose-dependent manner and did not impact sGnRH and cGnRH-II mRNA levels in vivo or in vitro. The inhibitory effect of E2 on sbGnRH expression was attenuated by the estrogen receptor (ER) broad-spectrum antagonist (fulvestrant) and the ERα-specific antagonist (methyl-piperidinopyrazole), respectively, implying that the feedback regulation on sbGnRH is mediated via ERα. This study provides a theoretical basis for the reproductive endocrinology of the spotted scat by studying GnRH.
Collapse
Affiliation(s)
- Hua-Pu Chen
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Fisheries College, Guangdong Ocean University, Zhanjiang, China
| | - Xue-Fan Cui
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Fisheries College, Guangdong Ocean University, Zhanjiang, China
| | - Yao-Rong Wang
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Fisheries College, Guangdong Ocean University, Zhanjiang, China
| | - Zhi-Yuan Li
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Fisheries College, Guangdong Ocean University, Zhanjiang, China
| | - Chang-Xu Tian
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Fisheries College, Guangdong Ocean University, Zhanjiang, China
| | - Dong-Neng Jiang
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Fisheries College, Guangdong Ocean University, Zhanjiang, China
| | - Chun-Hua Zhu
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Fisheries College, Guangdong Ocean University, Zhanjiang, China
| | - Yong Zhang
- State Key Laboratory of Biocontrol, and the Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, China
| | - Shui-Sheng Li
- State Key Laboratory of Biocontrol, and the Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, China.
| | - Guang-Li Li
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Fisheries College, Guangdong Ocean University, Zhanjiang, China.
| |
Collapse
|
8
|
Acharya KD, Nettles SA, Lichti CF, Warre-Cornish K, Polit LD, Srivastava DP, Denner L, Tetel MJ. Dopamine-induced interactions of female mouse hypothalamic proteins with progestin receptor-A in the absence of hormone. J Neuroendocrinol 2020; 32:e12904. [PMID: 33000549 PMCID: PMC7591852 DOI: 10.1111/jne.12904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 08/24/2020] [Accepted: 08/25/2020] [Indexed: 11/26/2022]
Abstract
Neural progestin receptors (PR) function in reproduction, neural development, neuroprotection, learning, memory and the anxiety response. In the absence of progestins, PR can be activated by dopamine (DA) in the rodent hypothalamus to elicit female sexual behaviour. The present study investigated mechanisms of DA activation of PR by testing the hypothesis that proteins from DA-treated hypothalami interact with PR in the absence of progestins. Ovariectomised, oestradiol-primed mice were infused with a D1-receptor agonist, SKF38393 (SKF), into the third ventricle 30 minutes prior to death. Proteins from SKF-treated hypothalami were pulled-down with glutathione S-transferase-tagged mouse PR-A or PR-B and the interactomes were analysed by mass spectrometry. The largest functional group to interact with PR-A in a DA-dependent manner was synaptic proteins. To test the hypothesis that DA activation of PR regulates synaptic proteins, we developed oestradiol-induced PR-expressing hypothalamic-like neurones derived from human-induced pluripotent stem cells (hiPSCs). Similar to progesterone (P4), SKF treatment of hiPSCs increased synapsin1/2 expression. This SKF-dependent effect was blocked by the PR antagonist RU486, suggesting that PR are necessary for this DA-induced increase. The second largest DA-dependent PR-A protein interactome comprised metabolic regulators involved in glucose metabolism, lipid synthesis and mitochondrial energy production. Interestingly, hypothalamic proteins interacted with PR-A, but not PR-B, in an SKF-dependent manner, suggesting that DA promotes the interaction of multiple hypothalamic proteins with PR-A. These in vivo and in vitro results indicate novel mechanisms by which DA can differentially activate PR isoforms in the absence of P4 and provide a better understanding of ligand-independent PR activation in reproductive, metabolic and mental health disorders in women.
Collapse
Affiliation(s)
| | | | - Cheryl F. Lichti
- Department of Pathology and Immunology, Washington University School of Medicine, 660 S Euclid Ave, St. Louis, MO 63110
| | - Katherine Warre-Cornish
- Department of Basic and Clinical Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry Psychology and Neuroscience, King’s College London, London, SE5 8AF, UK
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, UK
| | - Lucia Dutan Polit
- Department of Basic and Clinical Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry Psychology and Neuroscience, King’s College London, London, SE5 8AF, UK
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, UK
| | - Deepak P. Srivastava
- Department of Basic and Clinical Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry Psychology and Neuroscience, King’s College London, London, SE5 8AF, UK
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, UK
| | - Larry Denner
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555
| | - Marc J. Tetel
- Neuroscience Department, Wellesley College, Wellesley, MA 02481
| |
Collapse
|
9
|
Golovchenko O, Abramova M, Ponomarenko I, Reshetnikov E, Aristova I, Polonikov A, Dvornyk V, Churnosov M. Functionally significant polymorphisms of ESR1and PGR and risk of intrauterine growth restriction in population of Central Russia. Eur J Obstet Gynecol Reprod Biol 2020; 253:52-57. [PMID: 32777541 DOI: 10.1016/j.ejogrb.2020.07.045] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/21/2020] [Accepted: 07/24/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE This study aimed to investigate the role ofESR1 and PGR gene polymorphisms in development of intrauterine growth restriction (IUGR) among Russian women in Central Russia. STUDY DESIGN This case-control study recruited a total of 520 women in the third trimester of pregnancy, including 196 IUGR patients and 324 controls. The participants were unrelated women of self-reported Russian ethnicity. Participants were genotyped at 4 functionally significant polymorphisms of theESR1 (rs2234693, rs9340799) and the PGR (rs484389, rs1042838) genes. The association analysis was performed using logistic regression. Two polymorphisms, which were associated with IUGR, and 26 polymorphisms linked to them (r2≥0.6) were analyzed for their functional significance in silico. RESULTS Haplotype TG of loci rs2234693-rs9340799ESR1 (OR = 1.94, рperm = 0.006) was associated with an increased risk of IUGR. Allele T of rs2234693 decreases expression of ESR1 in thyroid gland, allele T of rs2234693 and allele G of rs9340799 increase affinity to eight transcription factors (AP-4, HEN1, E2A, LBP-1, RP58, LUN, Ets and Hand). The loci that are linked (r2≥0.6) to the IUGR-associated SNPs, have the cis-eQTL value (expression ESR1 in thyroid gland) and showed their regulatory effects in organs and tissues related to pathogenesis of IUGR. CONCLUSION Haplotype TG defined by polymorphisms rs2234693-rs9340799 of theESR1 gene is associated with the development of IUGR in Russian women from Central Russia.
Collapse
Affiliation(s)
- Oleg Golovchenko
- Department of Medical Biological Disciplines, Belgorod State University, 308015 Belgorod, Russia
| | - Maria Abramova
- Department of Medical Biological Disciplines, Belgorod State University, 308015 Belgorod, Russia
| | - Irina Ponomarenko
- Department of Medical Biological Disciplines, Belgorod State University, 308015 Belgorod, Russia
| | - Evgeny Reshetnikov
- Department of Medical Biological Disciplines, Belgorod State University, 308015 Belgorod, Russia.
| | - Inna Aristova
- Department of Medical Biological Disciplines, Belgorod State University, 308015 Belgorod, Russia
| | - Alexey Polonikov
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 305041 Kursk, Russia
| | - Volodymyr Dvornyk
- Department of Life Sciences, College of Science and General Studies, Alfaisal University, 11533 Riyadh, Saudi Arabia
| | - Mikhail Churnosov
- Department of Medical Biological Disciplines, Belgorod State University, 308015 Belgorod, Russia
| |
Collapse
|
10
|
Suetomi Y, Tatebayashi R, Sonoda S, Munetomo A, Matsuyama S, Inoue N, Uenoyama Y, Takeuchi Y, Tsukamura H, Ohkura S, Matsuda F. Establishment of immortalised cell lines derived from female Shiba goat KNDy and GnRH neurones. J Neuroendocrinol 2020; 32:e12857. [PMID: 32432378 DOI: 10.1111/jne.12857] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 04/10/2020] [Accepted: 04/20/2020] [Indexed: 12/27/2022]
Abstract
Kisspeptin plays a critical role in governing gonadotrophin-releasing hormone (GnRH)/gonadotrophin secretion and subsequent reproductive function in mammals. The hypothalamic arcuate nucleus (ARC) kisspeptin neurones, which co-express neurokinin B (NKB) and dynorphin A (Dyn) and are referred to as KNDy neurones, are considered to be involved in GnRH generation. The present study aimed to establish cell lines derived from goat KNDy and GnRH neurones. Primary-cultured cells of female Shiba goat foetal hypothalamic ARC and preoptic area (POA) tissues were immortalised with the infection of lentivirus containing the simian virus 40 large T-antigen gene. Clones of the immortalised cells were selected by the gene expression of a neuronal marker, and then the neurone-derived cell clones were further selected by the gene expression of KNDy or GnRH neurone markers. As a result, we obtained a KNDy neurone cell line (GA28) from the ARC, as well as two GnRH neurone cell lines (GP11 and GP31) from the POA. Immunocytochemistry revealed the expression of kisspeptin, NKB and Dyn in GA28 cells, as well as GnRH in GP11 and GP31 cells. GnRH secretion from GP11 and GP31 cells into the media was confirmed by an enzyme immunoassay. Moreover, kisspeptin challenge increased intracellular Ca2+ levels in subsets of both GP11 and GP31 cells. Kisspeptin mRNA expression in GA28 cells, which expressed the oestrogen receptor alpha gene, was significantly reduced by 17β-oestradiol treatment. Furthermore, the transcriptional core promoter and repressive regions of the goat NKB gene were detected using GA28 cells. In conclusion, we have established goat KNDy and GnRH neurone cell lines that could be used to analyse molecular and cellular mechanisms regulating KNDy and GnRH neurones in vitro, facilitating the clarification of reproductive neuroendocrine mechanisms in ruminants.
Collapse
Affiliation(s)
- Yuta Suetomi
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Ryoki Tatebayashi
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Shuhei Sonoda
- Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Arisa Munetomo
- Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Shuichi Matsuyama
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Naoko Inoue
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Yoshihisa Uenoyama
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Yukari Takeuchi
- Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Hiroko Tsukamura
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Satoshi Ohkura
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Fuko Matsuda
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
- Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
11
|
Inserra PIF, Charif SE, Fidel V, Giacchino M, Schmidt AR, Villarreal FM, Proietto S, Cortasa SA, Corso MC, Gariboldi MC, Leopardo NP, Fraunhoffer NA, Di Giorgio NP, Lux-Lantos VA, Halperin J, Vitullo AD, Dorfman VB. The key action of estradiol and progesterone enables GnRH delivery during gestation in the South American plains vizcacha, Lagostomus maximus. J Steroid Biochem Mol Biol 2020; 200:105627. [PMID: 32070756 DOI: 10.1016/j.jsbmb.2020.105627] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 02/07/2020] [Accepted: 02/09/2020] [Indexed: 11/23/2022]
Abstract
The South American plains vizcacha, Lagostomus maximus, is the only mammal described so far that shows expression of estrogen receptors (ERs) and progesterone receptors (PRs) in gonadotropin-releasing hormone (GnRH) neurons. This animal therefore constitutes an exceptional model for the study of the effect of steroid hormones on the modulation of the hypothalamic-pituitary-ovarian (HPO) axis. By using both in vivo and ex vivo approaches, we have found that pharmacological doses of progesterone (P4) and estradiol (E2) produced an inhibition in the expression of hypothalamic GnRH, while physiological doses produced a differential effect on the pulsatile release frequency or genomic expression of GnRH. Our ex vivo experiment indicates that a short-term effect of E2 modulates the frequency of GnRH release pattern that would be associated with membrane ERs. On the other hand, our in vivo approach suggests that a long-term effect of E2, acting through the classical nuclear ERs-PRs pathway, would produce the modification of GnRH mRNA expression during the GnRH pre-ovulatory surge. Particularly, P4 induced a rise in GnRH mRNA expression and protein release with a decrease in its release frequency. These results suggest different levels of action of steroid hormones on GnRH modulation. We conclude that the fine action of E2 and P4 constitute the key factor to enable the hypothalamic activity during the pregnancy of this mammal.
Collapse
Affiliation(s)
- Pablo I F Inserra
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina(2); Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Santiago E Charif
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina(2); Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Victoria Fidel
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina(2)
| | - Mariela Giacchino
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina(2); Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Alejandro R Schmidt
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina(2); Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Federico M Villarreal
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina(2)
| | - Sofía Proietto
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina(2); Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Santiago A Cortasa
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina(2); Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - María C Corso
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina(2); Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - María C Gariboldi
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina(2); Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Noelia P Leopardo
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina(2); Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Nicolás A Fraunhoffer
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina(2); Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Noelia P Di Giorgio
- Laboratorio de Neuroendocrinología, Instituto de Biología y Medicina Experimental (IByME)-CONICET, Ciudad Autónoma de Buenos Aires, Argentina
| | - Victoria A Lux-Lantos
- Laboratorio de Neuroendocrinología, Instituto de Biología y Medicina Experimental (IByME)-CONICET, Ciudad Autónoma de Buenos Aires, Argentina
| | - Julia Halperin
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina(2); Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Alfredo D Vitullo
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina(2); Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Verónica B Dorfman
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina(2); Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina.
| |
Collapse
|
12
|
Novaira HJ, Negron AL, Graceli JB, Capellino S, Schoeffield A, Hoffman GE, Levine JE, Wolfe A, Wondisford FE, Radovick S. Impairments in the reproductive axis of female mice lacking estrogen receptor β in GnRH neurons. Am J Physiol Endocrinol Metab 2018; 315:E1019-E1033. [PMID: 30040478 PMCID: PMC6293171 DOI: 10.1152/ajpendo.00173.2018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/02/2018] [Accepted: 07/21/2018] [Indexed: 12/24/2022]
Abstract
The effect of estrogen on the differentiation and maintenance of reproductive tissues is mediated by two nuclear estrogen receptors (ERs), ERα, and ERβ. Lack of functional ERα and ERβ genes in vivo significantly affects reproductive function; however, the target tissues and signaling pathways in the hypothalamus are not clearly defined. Here, we describe the generation and reproductive characterization of a complete-ERβ KO (CERβKO) and a GnRH neuron-specific ERβKO (GERβKO) mouse models. Both ERβKO mouse models displayed a delay in vaginal opening and first estrus. Hypothalamic gonadotropin-releasing hormone (GnRH) mRNA expression levels in both ERβKO mice were similar to control mice; however female CERβKO and GERβKO mice had lower basal and surge serum gonadotropin levels. Although a GnRH stimulation test in both female ERβKO models showed preserved gonadotropic function in the same animals, a kisspeptin stimulation test revealed an attenuated response by GnRH neurons, suggesting a role for ERβ in normal GnRH neuron function. No alteration in estrogen-negative feedback was observed in either ERβKO mouse models after ovariectomy and estrogen replacement. Further, abnormal development of ovarian follicles with low serum estradiol levels and impairment of fertility were observed in both ERβKO mouse models. In male ERβKO mice, no differences in the timing of pubertal onset or serum luteinizing hormone and follicle-stimulating hormone levels were observed as compared with controls. Taken together, these data provide in vivo evidence for a role of ERβ in GnRH neurons in modulating puberty and reproduction, specifically through kisspeptin responsiveness in the female hypothalamic-pituitary-gonadal axis.
Collapse
Affiliation(s)
- Horacio J Novaira
- Department of Pediatrics, Rutgers-Robert Wood Johnson Medical School , New Brunswick, New Jersey
| | - Ariel L Negron
- Department of Pediatrics, Rutgers-Robert Wood Johnson Medical School , New Brunswick, New Jersey
| | - Jones B Graceli
- Department of Morphology, Federal University of Espirito Santo , Vitoria , Brazil
| | - Silvia Capellino
- IfADo-Leibniz Research Centre for Working Environment and Human Factors, Department of Immunology , Dortmund , Germany
| | | | - Gloria E Hoffman
- Department of Biology, Morgan State University , Baltimore, Maryland
| | - Jon E Levine
- Department of Neuroscience, University of Wisconsin , Madison, Wisconsin
| | - Andrew Wolfe
- Department of Pediatrics, Division of Endocrinology, Johns Hopkins University School of Medicine , Baltimore, Maryland
| | - Fredric E Wondisford
- Department of Medicine, Rutgers-Robert Wood Johnson Medical School , New Brunswick, New Jersey
| | - Sally Radovick
- Department of Pediatrics, Rutgers-Robert Wood Johnson Medical School , New Brunswick, New Jersey
| |
Collapse
|
13
|
Loutchanwoot P, Vortherms T. Effects of puerarin on estrogen-regulated gene expression in gonadotropin-releasing hormone pulse generator of ovariectomized rats. Steroids 2018; 135:54-62. [PMID: 29733861 DOI: 10.1016/j.steroids.2018.05.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 04/28/2018] [Accepted: 05/02/2018] [Indexed: 11/28/2022]
Abstract
Effects of puerarin on the hypothalamic gonadotropin-releasing hormone (GnRH) pulse generator function is investigated, for the first time, in ovariectomized rats at the level of mRNA expression of estrogen-responsive genes, e.g., estrogen receptor (ER), GnRH and its receptor (GnRHR). Rats were treated orally for 90 days either with a soy-free diet containing two different doses of puerarin (low dose of 600 mg/kg and high dose of 3000 mg/kg) or estradiol benzoate (E2B) at either low dose (4.3 mg/kg) or high dose (17.3 mg/kg). Levels of mRNA expression in the medial preoptic area/anterior hypothalamus (MPOA/AH), mediobasal hypothalamus/median eminence (MBH/ME) and adenohypophysis were measured by quantitative TaqMan® real-time RT-PCR. Plasma levels of luteinizing hormone (LH) and prolactin (PRL) were measured by radioimmunoassay. In the MPOA/AH, both puerarin and E2B decreased ERα mRNA levels without any significant changes in ERβ and GnRH mRNA levels. Both puerarin and E2B did not significantly alter the expression levels of ERα, ERβ and GnRHR in the MBH/ME. E2B exerted significant effects on the down-regulation of adenohypophyseal GnRHR mRNA transcripts and serum LH levels. Puerarin did not cause significant changes in pituitary GnRHR mRNA transcripts and serum LH and PRL levels. This is the first study to demonstrate that in ovariectomized rat models of ovarian hormone deprivation, puerarin acted as a weak estrogen-active compound in the hypothalamic GnRH pulse generator through the downregulation of MPOA/AH ERα mRNA expression.
Collapse
Affiliation(s)
- Panida Loutchanwoot
- Department of Biology, Faculty of Science, Mahasarakham University, Khamriang Sub-district, Kantarawichai District, Mahasarakham Province 44150, Thailand.
| | - Tina Vortherms
- Department of Endocrinology, Faculty of Medicine, University Medical Center Göttingen, Robert-Koch-Strasse 40, D-37075 Göttingen, Germany
| |
Collapse
|
14
|
Park CJ, Chen G, Koo Y, Lin PCP, Cacioppo JA, Prohaska H, Ko CJ. Generation and characterization of an estrogen receptor alpha-iCre knock-in mouse. Genesis 2017; 55. [PMID: 29115049 DOI: 10.1002/dvg.23084] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 11/03/2017] [Accepted: 11/03/2017] [Indexed: 01/11/2023]
Abstract
Two estrogen receptors, ESR1 and ESR2, are responsible for the classical actions of estrogens in mammalian species. They display different spatiotemporal expression patterns and nonoverlapping functions in various tissues and physiological conditions. In this study, a novel knock-in mouse line that expresses codon-improved Cre recombinase (iCre) under regulation of the natural Esr1 promoter (Esr1-iCre) was developed. Functional characterization of iCre expression by crossing them with reporter lines (ROSA26-lacZ or Ai9-RFP) showed that iCre is faithfully expressed in Esr1-lineage cells. This novel transgenic mouse line will be a useful animal model for lineage-tracing Esr1-expressing cells, selective gene ablation in the Esr1-lineage cells and for generating global Esr1 knockout mice.
Collapse
Affiliation(s)
- Chan Jin Park
- Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana-Campaign, Illinois
| | - Guanglin Chen
- Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana-Campaign, Illinois
| | - Yongbum Koo
- School of Biological Sciences, Inje University, Gimhae, South Korea
| | - Po-Ching P Lin
- Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana-Campaign, Illinois
| | - Joseph A Cacioppo
- Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana-Campaign, Illinois
| | - Hailey Prohaska
- Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana-Campaign, Illinois
| | - CheMyong J Ko
- Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana-Campaign, Illinois
| |
Collapse
|
15
|
Charif SE, Inserra PIF, Schmidt AR, Di Giorgio NP, Cortasa SA, Gonzalez CR, Lux-Lantos V, Halperin J, Vitullo AD, Dorfman VB. Local production of neurostradiol affects gonadotropin-releasing hormone (GnRH) secretion at mid-gestation in Lagostomus maximus (Rodentia, Caviomorpha). Physiol Rep 2017; 5:5/19/e13439. [PMID: 29038356 PMCID: PMC5641931 DOI: 10.14814/phy2.13439] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 08/17/2017] [Indexed: 01/07/2023] Open
Abstract
Females of the South American plains vizcacha, Lagostomus maximus, show peculiar reproductive features such as massive polyovulation up to 800 oocytes per estrous cycle and an ovulatory process around mid‐gestation arising from the reactivation of the hypothalamic–hypophyseal–ovary (H.H.O.) axis. Estradiol (E2) regulates gonadotropin‐releasing hormone (GnRH) expression. Biosynthesis of estrogens results from the aromatization of androgens by aromatase, which mainly occurs in the gonads, but has also been described in the hypothalamus. The recently described correlation between GnRH and ERα expression patterns in the hypothalamus of the vizcacha during pregnancy, with coexpression in the same neurons of the medial preoptic area, suggests that hypothalamic synthesis of E2 may affect GnRH neurons and contribute with systemic E2 to modulate GnRH delivery during the gestation. To elucidate this hypothesis, hypothalamic expression and the action of aromatase on GnRH release were evaluated in female vizcachas throughout pregnancy. Aromatase and GnRH expression was increased significantly in mid‐pregnant and term‐pregnant vizcachas compared to early‐pregnant and nonpregnant females. In addition, aromatase and GnRH were colocalized in neurons of the medial preoptic area of the hypothalamus throughout gestation. The blockage of the negative feedback of E2 induced by the inhibition of aromatase resulted in a significant increment of GnRH‐secreted mass by hypothalamic explants. E2 produced in the same neurons as GnRH may drive intracellular E2 to higher levels than those obtained from systemic circulation alone. This may trigger for a prompt GnRH availability enabling H.H.O. activity at mid‐gestation with ovulation and formation of accessory corpora lutea with steroidogenic activity that produce the necessary progesterone to maintain gestation to term and guarantee the reproductive success.
Collapse
Affiliation(s)
- Santiago E Charif
- Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas CONICET, Ciudad Autónoma de Buenos Aires, Argentina
| | - Pablo I F Inserra
- Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas CONICET, Ciudad Autónoma de Buenos Aires, Argentina
| | - Alejandro R Schmidt
- Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas CONICET, Ciudad Autónoma de Buenos Aires, Argentina
| | - Noelia P Di Giorgio
- Consejo Nacional de Investigaciones Científicas y Técnicas CONICET, Ciudad Autónoma de Buenos Aires, Argentina.,Laboratorio de Neuroendocrinología, Instituto de Biología y Medicina Experimental, IByME-CONICET, Ciudad Autónoma de Buenos Aires, Argentina
| | - Santiago A Cortasa
- Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas CONICET, Ciudad Autónoma de Buenos Aires, Argentina
| | - Candela R Gonzalez
- Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas CONICET, Ciudad Autónoma de Buenos Aires, Argentina
| | - Victoria Lux-Lantos
- Consejo Nacional de Investigaciones Científicas y Técnicas CONICET, Ciudad Autónoma de Buenos Aires, Argentina.,Laboratorio de Neuroendocrinología, Instituto de Biología y Medicina Experimental, IByME-CONICET, Ciudad Autónoma de Buenos Aires, Argentina
| | - Julia Halperin
- Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas CONICET, Ciudad Autónoma de Buenos Aires, Argentina
| | - Alfredo Daniel Vitullo
- Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas CONICET, Ciudad Autónoma de Buenos Aires, Argentina
| | - Verónica B Dorfman
- Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina .,Consejo Nacional de Investigaciones Científicas y Técnicas CONICET, Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
16
|
ERα and GnRH co-localize in the hypothalamic neurons of the South American plains vizcacha, Lagostomus maximus (Rodentia, Caviomorpha). J Mol Histol 2017; 48:259-273. [DOI: 10.1007/s10735-017-9715-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 03/04/2017] [Indexed: 10/19/2022]
|
17
|
Voutsadakis IA. Hormone Receptors in Serous Ovarian Carcinoma: Prognosis, Pathogenesis, and Treatment Considerations. CLINICAL MEDICINE INSIGHTS-ONCOLOGY 2016; 10:17-25. [PMID: 27053923 PMCID: PMC4814131 DOI: 10.4137/cmo.s32813] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 02/09/2016] [Accepted: 02/11/2016] [Indexed: 12/15/2022]
Abstract
A few breakthroughs have been accomplished for the treatment of ovarian cancer, the most deadly gynecologic carcinoma, in the current era of targeted oncologic treatment. The estrogen receptor was the first target of such treatments with the introduction of tamoxifen four decades ago in breast cancer therapeutics. Attempts to duplicate the success of hormonal therapies in ovarian cancer met with mixed results, which may be due to an inferior degree of hormone dependency in this cancer. Alternatively, this may be due to the failure to clearly identify the subsets of ovarian cancer with hormone sensitivity. This article reviews the expression of hormone receptors by ovarian cancer cells, the prognostic value of these expressions, and their predictive capacity for response to hormonal agents. The possible ways ahead are briefly discussed.
Collapse
Affiliation(s)
- Ioannis A Voutsadakis
- Division of Medical Oncology, Department of Internal Medicine, Sault Area Hospital, Sault Ste Marie, ON, Canada.; Division of Clinical Sciences, Northern Ontario School of Medicine, Sudbury, ON, Canada
| |
Collapse
|
18
|
Effects of Huang Bai (Phellodendri Cortex) and Three Other Herbs on GnRH and GH Levels in GT1-7 and GH3 Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:9389028. [PMID: 26925153 PMCID: PMC4746385 DOI: 10.1155/2016/9389028] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 12/16/2015] [Indexed: 11/24/2022]
Abstract
The present study was to evaluate the effects of Huang Bai, Zhi Mu, Mai Ya, and Xia Ku Cao on hormone using the GT1–7 and GH3 cells. The GT1–7 and GH3 cell lines were incubated with DW; DMSO; and 30, 100, or 300 μg/mL of one of the four extract solutions in serum-free media for 24 hours. The MTT assay was performed to determine the cytotoxicity of the four herbs. The GT1–7 and GH3 cells were incubated in DW, estradiol (GT1–7 only), or noncytotoxic herb solutions in serum-free medium for 24 hours. A quantitative RT-PCR and western blot were performed to measure the GnRH expression in GT1–7 cells and GH expression in GH3 cells. Huang Bai, Zhi Mu, Xia Ku Cao, and Mai Ya inhibited the GnRH mRNA expression in GT1–7 cells, whereas Huang Bai enhanced GH mRNA expression in GH3 cells. Additionally, Xia Ku Cao inhibited GnRH protein expression in GT1–7 cells and Huang Bai promoted GH protein expression in GH3 cells. The findings suggest that Huang Bai can delay puberty by inhibiting GnRH synthesis in the hypothalamus while also accelerating growth by promoting GH synthesis and secretion in the pituitary.
Collapse
|
19
|
Treen AK, Luo V, Chalmers JA, Dalvi PS, Tran D, Ye W, Kim GL, Friedman Z, Belsham DD. Divergent Regulation of ER and Kiss Genes by 17β-Estradiol in Hypothalamic ARC Versus AVPV Models. Mol Endocrinol 2016; 30:217-33. [PMID: 26726951 DOI: 10.1210/me.2015-1189] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Kisspeptin (Kiss) and G-protein-coupled receptor (Gpr)54 have emerged as key regulators of reproduction. 17β-estradiol (E2)-mediated regulation of these neurons is nuclei specific, where anteroventral periventricular (AVPV) Kiss neurons are positively regulated by E2, whereas arcuate nucleus (ARC) neurons are inhibited. We have generated immortalized Kiss cell lines from male and female adult-derived murine hypothalamic primary culture, as well as cell lines from microdissected AVPV and ARC from female Kiss-green fluorescent protein (GFP) mice. All exhibit endogenous Kiss-1 expression, estrogen receptors (ER)s (ERα, ERβ, and Gpr30), as well as known markers of AVPV Kiss neurons in the mHypoA-50 and mHypoA-Kiss/GFP-4, vs markers of ARC Kiss neurons in the mHypoA-55 and the mHypoA-Kiss/GFP-3 lines. There was an increase in Kiss-1 mRNA expression at 24 hours in the AVPV lines and a repression of Kiss-1 mRNA at 4 hours in the ARC lines. An E2-mediated decrease in ERα mRNA expression at 24 hours in the AVPV cell lines was detected, and a significant decrease in Gpr30, ERα, and ERβ mRNA levels at 4 hours in the ARC cell lines was evident. ER agonists and antagonists determined the specific ERs responsible for mediating changes in gene expression. In the AVPV, ERα is required but not ERβ or GPR30, vs the ARC Kiss-expressing cell lines that require GPR30, and either ERα and/or ERβ. We determined cAMP response element-binding protein 1 was necessary for the down-regulation of Kiss-1 mRNA expression using small interfering RNA knockdown in the ARC cell model. These studies elucidate some of the molecular events involved in the differential E2-mediated regulation of unique and specific Kiss neuronal models.
Collapse
Affiliation(s)
- Alice K Treen
- Departments of Physiology (A.K.T., V.L., J.A.C., P.S.D., D.T., W.Y., G.L.K., Z.F., D.D.B.), Medicine (D.D.B.), and Obstetrics and Gynaecology (D.D.B.), University of Toronto, and Division of Cellular and Molecular Biology (D.D.B.), Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada M5S 1A8
| | - Vicky Luo
- Departments of Physiology (A.K.T., V.L., J.A.C., P.S.D., D.T., W.Y., G.L.K., Z.F., D.D.B.), Medicine (D.D.B.), and Obstetrics and Gynaecology (D.D.B.), University of Toronto, and Division of Cellular and Molecular Biology (D.D.B.), Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada M5S 1A8
| | - Jennifer A Chalmers
- Departments of Physiology (A.K.T., V.L., J.A.C., P.S.D., D.T., W.Y., G.L.K., Z.F., D.D.B.), Medicine (D.D.B.), and Obstetrics and Gynaecology (D.D.B.), University of Toronto, and Division of Cellular and Molecular Biology (D.D.B.), Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada M5S 1A8
| | - Prasad S Dalvi
- Departments of Physiology (A.K.T., V.L., J.A.C., P.S.D., D.T., W.Y., G.L.K., Z.F., D.D.B.), Medicine (D.D.B.), and Obstetrics and Gynaecology (D.D.B.), University of Toronto, and Division of Cellular and Molecular Biology (D.D.B.), Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada M5S 1A8
| | - Dean Tran
- Departments of Physiology (A.K.T., V.L., J.A.C., P.S.D., D.T., W.Y., G.L.K., Z.F., D.D.B.), Medicine (D.D.B.), and Obstetrics and Gynaecology (D.D.B.), University of Toronto, and Division of Cellular and Molecular Biology (D.D.B.), Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada M5S 1A8
| | - Wenqing Ye
- Departments of Physiology (A.K.T., V.L., J.A.C., P.S.D., D.T., W.Y., G.L.K., Z.F., D.D.B.), Medicine (D.D.B.), and Obstetrics and Gynaecology (D.D.B.), University of Toronto, and Division of Cellular and Molecular Biology (D.D.B.), Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada M5S 1A8
| | - Ginah L Kim
- Departments of Physiology (A.K.T., V.L., J.A.C., P.S.D., D.T., W.Y., G.L.K., Z.F., D.D.B.), Medicine (D.D.B.), and Obstetrics and Gynaecology (D.D.B.), University of Toronto, and Division of Cellular and Molecular Biology (D.D.B.), Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada M5S 1A8
| | - Zoey Friedman
- Departments of Physiology (A.K.T., V.L., J.A.C., P.S.D., D.T., W.Y., G.L.K., Z.F., D.D.B.), Medicine (D.D.B.), and Obstetrics and Gynaecology (D.D.B.), University of Toronto, and Division of Cellular and Molecular Biology (D.D.B.), Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada M5S 1A8
| | - Denise D Belsham
- Departments of Physiology (A.K.T., V.L., J.A.C., P.S.D., D.T., W.Y., G.L.K., Z.F., D.D.B.), Medicine (D.D.B.), and Obstetrics and Gynaecology (D.D.B.), University of Toronto, and Division of Cellular and Molecular Biology (D.D.B.), Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada M5S 1A8
| |
Collapse
|
20
|
Naulé L, Robert V, Parmentier C, Martini M, Keller M, Cohen-Solal M, Hardin-Pouzet H, Grange-Messent V, Franceschini I, Mhaouty-Kodja S. Delayed pubertal onset and prepubertal Kiss1 expression in female mice lacking central oestrogen receptor beta. Hum Mol Genet 2015; 24:7326-38. [PMID: 26464488 DOI: 10.1093/hmg/ddv430] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 10/07/2015] [Indexed: 01/02/2023] Open
Abstract
Ovarian oestradiol is essential for pubertal maturation and adult physiology of the female reproductive axis. It acts at central and peripheral sites through two main oestrogen receptors (ER) α and β. Here we investigate the role of ERβ on central effects of oestradiol, by generating a mouse line specifically lacking the ERβ gene in neuronal and glial cells. Central ERβ deletion delays the age at vaginal opening and first oestrous and reduces uterine weight without affecting body growth. Analysis of factors necessary for pubertal progression shows reduced levels of Kiss1 transcripts at postnatal (P) day 25 in the preoptic area, but not in the mediobasal hypothalamus (MBH) of mutant females. In agreement with these data, the number of kisspeptin-immunoreactive neurons was decreased by 57-72% in the three subdivisions of the rostral periventricular area of the third ventricle (RP3V), whereas the density of kisspeptin-immunoreactive fibres was unchanged in the arcuate nucleus of mutant mice. These alterations do not involve changes in ERα mRNAs in the preoptic area and protein levels in the RP3V. The number and distribution of GnRH-immunoreactive cells were unaffected, but gonadotropin-releasing hormone (GnRH) transcript levels were higher in the P25 preoptic area of mutants. At adulthood, mutant females have normal oestrous cyclicity, kisspeptin system and exhibit unaltered sexual behaviour. They display, however, reduced ovary weight and increased anxiety-related behaviour during the follicular phase. This argues for the specific involvement of central ERβ in the regulation of pubertal onset in female reproduction, possibly through prepubertal induction of kisspeptin expression in the RP3V.
Collapse
Affiliation(s)
- Lydie Naulé
- Neuroscience Paris Seine, Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche (UMR) S1130, Centre National de la Recherche Scientifique, UMR 8246, Université P. et M. Curie, Paris, France, Sorbonne Universités, Université P. et M. Curie UM CR18, Université Paris 06, Paris, France
| | - Vincent Robert
- Institut National de la Recherche Agronomique, UMR 85, Nouzilly, France, Centre National de la Recherche Scientifique, UMR 7247, Nouzilly, France, Université François Rabelais, Tours, France, Institut Français du Cheval et de l'Equitation, Nouzilly, France and
| | - Caroline Parmentier
- Neuroscience Paris Seine, Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche (UMR) S1130, Centre National de la Recherche Scientifique, UMR 8246, Université P. et M. Curie, Paris, France, Sorbonne Universités, Université P. et M. Curie UM CR18, Université Paris 06, Paris, France
| | - Mariangela Martini
- Institut National de la Recherche Agronomique, UMR 85, Nouzilly, France, Centre National de la Recherche Scientifique, UMR 7247, Nouzilly, France, Université François Rabelais, Tours, France, Institut Français du Cheval et de l'Equitation, Nouzilly, France and
| | - Matthieu Keller
- Institut National de la Recherche Agronomique, UMR 85, Nouzilly, France, Centre National de la Recherche Scientifique, UMR 7247, Nouzilly, France, Université François Rabelais, Tours, France, Institut Français du Cheval et de l'Equitation, Nouzilly, France and
| | - Martine Cohen-Solal
- Inserm U1132 and university Paris-Diderot, Hospital Lariboisière, Paris, France
| | - Hélène Hardin-Pouzet
- Neuroscience Paris Seine, Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche (UMR) S1130, Centre National de la Recherche Scientifique, UMR 8246, Université P. et M. Curie, Paris, France, Sorbonne Universités, Université P. et M. Curie UM CR18, Université Paris 06, Paris, France
| | - Valérie Grange-Messent
- Neuroscience Paris Seine, Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche (UMR) S1130, Centre National de la Recherche Scientifique, UMR 8246, Université P. et M. Curie, Paris, France, Sorbonne Universités, Université P. et M. Curie UM CR18, Université Paris 06, Paris, France
| | - Isabelle Franceschini
- Institut National de la Recherche Agronomique, UMR 85, Nouzilly, France, Centre National de la Recherche Scientifique, UMR 7247, Nouzilly, France, Université François Rabelais, Tours, France, Institut Français du Cheval et de l'Equitation, Nouzilly, France and
| | - Sakina Mhaouty-Kodja
- Neuroscience Paris Seine, Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche (UMR) S1130, Centre National de la Recherche Scientifique, UMR 8246, Université P. et M. Curie, Paris, France, Sorbonne Universités, Université P. et M. Curie UM CR18, Université Paris 06, Paris, France,
| |
Collapse
|
21
|
Abstract
Brain development is an organized, but constantly adaptive, process in which genetic and epigenetic signals allow neurons to differentiate, to migrate, and to develop correct connections. Gender specific prenatal sex hormone milieu participates in the dimorphic development of many neuronal networks. Environmental cues may interfere with these developmental programs, producing adverse outcomes. Bisphenol A (BPA), an estrogenic/antiandrogenic endocrine disruptor widely diffused in the environment, produces adverse effects at levels below the acceptable daily intake. This review analyzes the recent literature on the consequences of perinatal exposure to BPA environmental doses on the development of a dimorphic brain. The BPA interference with the development and function of the neuroendocrine hypothalamus and of the nuclei controlling energy balance, and with the hippocampal memory processing is also discussed. The detrimental action of BPA appears complex, involving different hormonal and epigenetic pathways activated, often in a dimorphic way, within clearcut susceptibility windows. To date, discrepancies in experimental approaches and in related outcomes make unfeasible to translate the available information into clear dose-response models for human risk assessment. Evaluation of BPA brain levels in relation to the appearance of adverse effects in future basic studies will certainly give better definition of the warning threshold for human health.
Collapse
Affiliation(s)
- P Negri-Cesi
- Department of Pharmacological and Biomolecular Sciences, Section of Biomedicine and Endocrinology, INBB Research Unit, Milano, Italy
| |
Collapse
|
22
|
Chason RJ, Kang JH, Gerkowicz SA, Dufau ML, Catt KJ, Segars JH. GnRH agonist reduces estrogen receptor dimerization in GT1-7 cells: evidence for cross-talk between membrane-initiated estrogen and GnRH signaling. Mol Cell Endocrinol 2015; 404:67-74. [PMID: 25619861 PMCID: PMC4590284 DOI: 10.1016/j.mce.2015.01.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 01/18/2015] [Accepted: 01/19/2015] [Indexed: 12/27/2022]
Abstract
17β-estradiol (E2), a key participant on the initiation of the LH surge, exerts both positive and negative feedback on GnRH neurons. We sought to investigate potential interactions between estrogen receptors alpha (ERα) and beta (ERβ) and gonadotropin releasing hormone receptor (GnRH-R) in GT1-7 cells. Radioligand binding studies demonstrated a significant decrease in saturation E2 binding in cells treated with GnRH agonist. Conversely, there was a significant reduction in GnRH binding in GT1-7 cells treated with E2. In BRET(1) experiments, ERα-ERα dimerization was suppressed in GT1-7 cells treated with GnRH agonist (p < 0.05). There was no evidence of direct interaction between ERs and GnRH-R. This study provides the first evidence of reduced ERα homodimerization by GnRH agonist. Collectively, these findings demonstrate significant cross-talk between membrane-initiated GnRH and E2 signaling in GT1-7 cells.
Collapse
Affiliation(s)
- Rebecca J Chason
- Program in Reproductive and Adult Endocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, 10 CRC, Room 1E-3140, 10 Center Drive, MSC 1109, Bethesda, MD 20892-1109, USA
| | - Jung-Hoon Kang
- Section on Molecular Endocrinology, Program on Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-4510, USA
| | - Sabrina A Gerkowicz
- Department of Obstetrics and Gynecology, University of Miami, 1611 NW 12th Ave, Miami, FL 33136, USA
| | - Maria L Dufau
- Section on Molecular Endocrinology, Program on Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-4510, USA
| | - Kevin J Catt
- Endocrinology and Reproduction Research Branch, Section on Hormonal Regulation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - James H Segars
- Program in Reproductive and Adult Endocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, 10 CRC, Room 1E-3140, 10 Center Drive, MSC 1109, Bethesda, MD 20892-1109, USA.
| |
Collapse
|
23
|
Prathibha Y, Murugananthkumar R, Rajakumar A, Laldinsangi C, Sudhakumari CC, Mamta SK, Dutta-Gupta A, Senthilkumaran B. Gene expression analysis in gonads and brain of catfish Clarias batrachus after the exposure of malathion. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2014; 102:210-219. [PMID: 24468447 DOI: 10.1016/j.ecoenv.2013.12.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 12/27/2013] [Accepted: 12/28/2013] [Indexed: 06/03/2023]
Abstract
Pesticides like malathion have the potential to disrupt development and reproduction of aquatic organisms including fishes. To investigate the likely consequences of malathion exposure at low doses in juvenile catfish, Clarias batrachus, we studied the expression pattern of genes encoding certain transcription factors, activin A, sex steroid or orphan nuclear receptors and steroidogenic enzymes which are known to be involved in gonadal development along with histological changes. To compare further, we also analyzed certain brain specific genes related to gonadal axis. Fifty days post hatch catfish fingerlings were exposed continuously to 1 and 10 µg/L of malathion for 21 days. Results from these experiments indicated that transcript levels of various genes were altered by the treatments, which may further affect the gonadal development either directly or indirectly through brain. Histological analysis revealed slow progression of spermatogenesis in testis, while in ovary, the oil droplet oocytes were found to be higher after treatment (10 µg/L). Our findings revealed that the exposure of malathion, even at low doses, hinder or modulate early gonadal development differentially by targeting gene expression pattern of transcription factors, activin A, sex steroid or orphan nuclear receptors and steroidogenic enzymes with an evidence on histological changes. Further, some of the genes showed differential expression at the level of brain in male and female sex after the exposure of malathion.
Collapse
Affiliation(s)
- Y Prathibha
- Department of Animal Sciences, School of Life Sciences-Centre for Advanced Studies, University of Hyderabad, PO Central University, Hyderabad-500046, Andhra Pradesh, India
| | - R Murugananthkumar
- Department of Animal Sciences, School of Life Sciences-Centre for Advanced Studies, University of Hyderabad, PO Central University, Hyderabad-500046, Andhra Pradesh, India
| | - A Rajakumar
- Department of Animal Sciences, School of Life Sciences-Centre for Advanced Studies, University of Hyderabad, PO Central University, Hyderabad-500046, Andhra Pradesh, India
| | - C Laldinsangi
- Department of Animal Sciences, School of Life Sciences-Centre for Advanced Studies, University of Hyderabad, PO Central University, Hyderabad-500046, Andhra Pradesh, India
| | - C C Sudhakumari
- Department of Animal Sciences, School of Life Sciences-Centre for Advanced Studies, University of Hyderabad, PO Central University, Hyderabad-500046, Andhra Pradesh, India
| | - S K Mamta
- Department of Animal Sciences, School of Life Sciences-Centre for Advanced Studies, University of Hyderabad, PO Central University, Hyderabad-500046, Andhra Pradesh, India
| | - A Dutta-Gupta
- Department of Animal Sciences, School of Life Sciences-Centre for Advanced Studies, University of Hyderabad, PO Central University, Hyderabad-500046, Andhra Pradesh, India
| | - B Senthilkumaran
- Department of Animal Sciences, School of Life Sciences-Centre for Advanced Studies, University of Hyderabad, PO Central University, Hyderabad-500046, Andhra Pradesh, India.
| |
Collapse
|
24
|
Jayes FL, Burns KA, Rodriguez KF, Kissling GE, Korach KS. The naturally occurring luteinizing hormone surge is diminished in mice lacking estrogen receptor Beta in the ovary. Biol Reprod 2014; 90:24. [PMID: 24337314 DOI: 10.1095/biolreprod.113.113316] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Female ESR2-null mice (betaERKO) display defects in ovarian function and are subfertile. Follicular maturation is impaired and explains smaller litters, but betaERKO also produce fewer litters, which may be partially due to inadequate ovulatory signals. To test this, the amplitude and timing of the naturally occurring luteinizing hormone (LH) surge was measured in individual intact betaERKO and wild-type (WT) mice. Vaginal cytology was evaluated daily, and blood samples were taken from mice in proestrus. The amplitude of the LH surge was severely blunted in betaERKO mice compared to WT, but pituitary LH levels revealed no differences. The betaERKO mice did not produce a preovulatory estradiol surge. To determine if the smaller LH surges and the reduced number of litters in betaERKO were due to the lack of ESR2 in the hypothalamic-pituitary axis or due to the absence of ESR2 in the ovary, ovaries were transplanted from WT into betaERKO mice and vice versa. The size of the LH surge was reduced only in mice lacking ESR2 within the ovary, and these mice had fewer litters. Fertility and size of the LH surge were rescued in betaERKO mice receiving a WT ovary. These data provide the first experimental evidence that the LH surge is impaired in betaERKO females and may contribute to their reduced fertility. ESR2 is not necessary within the pituitary and hypothalamus for the generation of a normal LH surge and for normal fertility, but ESR2 is essential within the ovary to provide proper signals.
Collapse
Affiliation(s)
- Friederike L Jayes
- Receptor Biology Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | | | | | | | | |
Collapse
|
25
|
Heublein S, Mayr D, Vrekoussis T, Friese K, Hofmann SS, Jeschke U, Lenhard M. The G-protein coupled estrogen receptor (GPER/GPR30) is a gonadotropin receptor dependent positive prognosticator in ovarian carcinoma patients. PLoS One 2013; 8:e71791. [PMID: 23951246 PMCID: PMC3739730 DOI: 10.1371/journal.pone.0071791] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 07/02/2013] [Indexed: 11/18/2022] Open
Abstract
Follicle stimulating hormone receptor (FSHR) and luteinizing hormone receptor (LHCGR) were demonstrated to impact upon survival of patients suffering from epithelial ovarian cancer (EOC). Though structure wise the G-protein coupled estrogen receptor (GPER/GPR30) is related to FSHR/LHCGR, its prognostic impact in EOC remains controversial. We recently found that FSHR negative patients represent a specific EOC subgroup that may behave differently in respect to both treatment response and prognosis. Hence, the current study aimed to analyze how GPER may interact with the FSHR/LHCGR system in EOC and whether the prognostic significance of GPER in EOC cases (n = 151) may be dependent on the FSHR/LHCGR immunophenotype of the tumor. Ovarian cancer cell lines were used to study how FSH and LH regulate GPER and whether GPER activation differentially affects in vitro cell proliferation in presence/absence of activated FSHR/LHCGR. In EOC tissue, GPER correlated with FSHR/LHCGR and was related to prolonged overall survival only in FSHR/LHCGR negative patients. Although GPER was found to be specifically induced by LH/FSH, GPER agonists (4-Hydroxy-Tamoxifen, G1) reduced EOC cell proliferation only in case of LH/FSH unstimulated pathways. To the same direction, only patients characterized as LHCGR/FSHR negative seem to gain from GPER in terms of survival. Our combined tissue and in vitro results support thus the hypothesis that GPER activation could be of therapeutic benefit in LHCGR/FSHR negative EOC patients. Further studies are needed to evaluate the impact of GPER activation on a clinical scheme.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Ovarian Epithelial
- Cell Line, Tumor
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Middle Aged
- Neoplasm Staging
- Neoplasms, Glandular and Epithelial/drug therapy
- Neoplasms, Glandular and Epithelial/genetics
- Neoplasms, Glandular and Epithelial/mortality
- Neoplasms, Glandular and Epithelial/pathology
- Ovarian Neoplasms/drug therapy
- Ovarian Neoplasms/genetics
- Ovarian Neoplasms/mortality
- Ovarian Neoplasms/pathology
- Prognosis
- Receptor Cross-Talk/drug effects
- Receptors, Estrogen/agonists
- Receptors, Estrogen/genetics
- Receptors, Estrogen/metabolism
- Receptors, FSH/genetics
- Receptors, FSH/metabolism
- Receptors, G-Protein-Coupled/agonists
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Receptors, LH/genetics
- Receptors, LH/metabolism
- Retrospective Studies
- Selective Estrogen Receptor Modulators/pharmacology
- Signal Transduction
- Survival Analysis
- Tamoxifen/analogs & derivatives
- Tamoxifen/pharmacology
Collapse
Affiliation(s)
- Sabine Heublein
- Department of Obstetrics and Gynaecology, Campus Innenstadt, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Doris Mayr
- Department of Pathology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Thomas Vrekoussis
- Department of Obstetrics and Gynaecology, Campus Innenstadt, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Klaus Friese
- Department of Obstetrics and Gynaecology, Campus Innenstadt, Ludwig-Maximilians-University of Munich, Munich, Germany
- Department of Pathology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Simone S. Hofmann
- Department of Obstetrics and Gynaecology, Campus Innenstadt, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Udo Jeschke
- Department of Obstetrics and Gynaecology, Campus Innenstadt, Ludwig-Maximilians-University of Munich, Munich, Germany
- * E-mail:
| | - Miriam Lenhard
- Department of Obstetrics and Gynaecology, Campus Grosshadern, Ludwig-Maximilians-University of Munich, Munich, Germany
| |
Collapse
|
26
|
Larco DO, Semsarzadeh NN, Cho-Clark M, Mani SK, John Wu T. The Novel Actions of the Metabolite GnRH-(1-5) are Mediated by a G Protein-Coupled Receptor. Front Endocrinol (Lausanne) 2013; 4:83. [PMID: 23847594 PMCID: PMC3703583 DOI: 10.3389/fendo.2013.00083] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 06/24/2013] [Indexed: 12/31/2022] Open
Abstract
The gonadotropin-releasing hormone (GnRH) was originally isolated from the mammalian hypothalamus for its role as the primary regulator of reproductive function. Since its discovery, GnRH has also been shown to be located in non-hypothalamic tissues and is known to have diverse functions. Although the regulation of GnRH synthesis and release has been extensively studied, there is additional evidence to suggest that the processing of GnRH to the metabolite GnRH-(1-5) represents another layer of regulation. The focus of this review will be on the current evidence for the action of the pentapeptide metabolite GnRH-(1-5) in regulating cellular migration. We discuss the potential role of GnRH-(1-5) in regulating GnRH neuronal migration during development. Furthermore, we demonstrate these actions are mediated by the activation of a G protein-coupled receptor. Our findings suggest that GnRH-(1-5) may play a developmental function in addition to regulating developing cells.
Collapse
Affiliation(s)
- Darwin Omar Larco
- Program in Molecular and Cellular Biology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Nina Nashat Semsarzadeh
- Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Madelaine Cho-Clark
- Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Shaila K. Mani
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - T. John Wu
- Program in Molecular and Cellular Biology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- *Correspondence: T. John Wu, Department of Obstetrics and Gynecology, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA e-mail:
| |
Collapse
|
27
|
Parhar I, Ogawa S, Kitahashi T. RFamide peptides as mediators in environmental control of GnRH neurons. Prog Neurobiol 2012; 98:176-96. [DOI: 10.1016/j.pneurobio.2012.05.011] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2011] [Revised: 05/10/2012] [Accepted: 05/11/2012] [Indexed: 11/25/2022]
|
28
|
XU YONGJIANG, LIU XUEZHOU, LIAO MEIJIE, WANG HANPING, WANG QINGYIN. Molecular Cloning and Differential Expression of Three GnRH Genes during Ovarian Maturation of Spotted Halibut, Verasper variegatus. ACTA ACUST UNITED AC 2012; 317:434-46. [DOI: 10.1002/jez.1736] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Revised: 02/14/2012] [Accepted: 04/03/2012] [Indexed: 11/07/2022]
Affiliation(s)
| | - XUE-ZHOU LIU
- Yellow Sea Fisheries Research Institute; Chinese Academy of Fishery Sciences; Qingdao; Shandong; China
| | - MEI-JIE LIAO
- Yellow Sea Fisheries Research Institute; Chinese Academy of Fishery Sciences; Qingdao; Shandong; China
| | - HAN-PING WANG
- Aquaculture Genetics and Breeding Laboratory; The Ohio State University South Centers; Piketon; Ohio
| | - QING-YIN WANG
- Yellow Sea Fisheries Research Institute; Chinese Academy of Fishery Sciences; Qingdao; Shandong; China
| |
Collapse
|
29
|
Su C, Rybalchenko N, Schreihofer DA, Singh M, Abbassi B, Cunningham RL. Cell Models for the Study of Sex Steroid Hormone Neurobiology. ACTA ACUST UNITED AC 2012; S2. [PMID: 22860237 DOI: 10.4172/2157-7536.s2-003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
To date many aspects of neurons and glia biology remain elusive, due in part to the cellular and molecular complexity of the brain. In recent decades, cell models from different brain areas have been established and proven invaluable toward understanding this complexity. In the field of steroid hormone neurobiology, an important question is: what is the profile of steroid hormone receptor expression in these specific cell lines? Currently, a clear summary of such receptor profiling is lacking. For this reason, we summarized in this review the expression of estrogen, progesterone, and androgen receptors in several widely used cell lines (glial and neuronal) derived from the forebrain and midbrain, based on our own data and that from the literature. Such information will aid in the selection of specific cell lines used to test hypotheses related to the biology of estrogens, progestins, and/or androgens.
Collapse
Affiliation(s)
- Chang Su
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, Fort Worth, TX 76107 USA
| | | | | | | | | | | |
Collapse
|
30
|
Radovick S, Levine JE, Wolfe A. Estrogenic regulation of the GnRH neuron. Front Endocrinol (Lausanne) 2012; 3:52. [PMID: 22654870 PMCID: PMC3356008 DOI: 10.3389/fendo.2012.00052] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Accepted: 03/16/2012] [Indexed: 11/17/2022] Open
Abstract
Reproductive function is regulated by the secretion of luteinizing hormone (LH) and follicle-stimulating hormone from the pituitary and the steroid hormones from the gonads. The dynamic changes in the levels of the reproductive hormones regulate secondary sex characteristics, gametogenesis, cellular function, and behavior. Hypothalamic GnRH neurons, with cell bodies located in the basal hypothalamus, represent the final common pathway for neuronally derived signals to the pituitary. As such, they serve as integrators of a dizzying array of signals including sensory inputs mediating information about circadian, seasonal, behavioral, pheromonal, and emotional cues. Additionally, information about peripheral physiological function may also be included in the integrative signal to the GnRH neuron. These signals may communicate information about metabolic status, disease, or infection. Gonadal steroid hormones arguably exert the most important effects on GnRH neuronal function. In both males and females, the gonadal steroid hormones exert negative feedback regulation on axis activity at both the level of the pituitary and the hypothalamus. These negative feedback loops regulate homeostasis of steroid hormone levels. In females, a cyclic reversal of estrogen feedback produces a positive feedback loop at both the hypothalamic and pituitary levels. Central positive feedback results in a dramatic increase in GnRH secretion (Moenter et al., 1992; Xia et al., 1992; Clarke, 1993; Sisk et al., 2001). This is coupled with an increase in pituitary sensitivity to GnRH (Savoy-Moore et al., 1980; Turzillo et al., 1995), which produces the massive surge in secretion of LH that triggers ovulation. While feedback regulation of the axis in males is in part mediated by estrogen receptors (ER), there is not a clear consensus as to the relative role of ER versus AR signaling in males (Lindzey et al., 1998; Wersinger et al., 1999). Therefore, this review will focus on estrogenic signaling in the female.
Collapse
Affiliation(s)
- Sally Radovick
- Department of Pediatrics, Johns Hopkins University School of MedicineBaltimore, MD, USA
| | - Jon E. Levine
- Wisconsin National Primate Research CenterMadison, WI, USA
| | - Andrew Wolfe
- Department of Pediatrics, Johns Hopkins University School of MedicineBaltimore, MD, USA
- *Correspondence: Andrew Wolfe, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA. e-mail:
| |
Collapse
|
31
|
Abstract
Oestradiol exerts a profound influence upon multiple brain circuits. For the most part, these effects are mediated by oestrogen receptor (ER)α. We review here the roles of ERβ, the other ER isoform, in mediating rodent oestradiol-regulated anxiety, aggressive and sexual behaviours, the control of gonadotrophin secretion, and adult neurogenesis. Evidence exists for: (i) ERβ located in the paraventricular nucleus underpinning the suppressive influence of oestradiol on the stress axis and anxiety-like behaviour; (ii) ERβ expressed in gonadotrophin-releasing hormone neurones contributing to oestrogen negative-feedback control of gonadotrophin secretion; (iii) ERβ controlling the offset of lordosis behaviour; (iv) ERβ suppressing aggressive behaviour in males; (v) ERβ modulating responses to social stimuli; and (vi) ERβ in controlling adult neurogenesis. This review highlights two major themes; first, ERβ and ERα are usually tightly inter-related in the oestradiol-dependent control of a particular brain function. For example, even though oestradiol feedback to control reproduction occurs principally through ERα-dependent mechanisms, modulatory roles for ERβ also exist. Second, the roles of ERα and ERβ within a particular neural network may be synergistic or antagonistic. Examples of the latter include the role of ERα to enhance, and ERβ to suppress, anxiety-like and aggressive behaviours. Splice variants such as ERβ2, acting as dominant negative receptors, are of further particular interest because their expression levels may reflect preceeding oestradiol exposure of relevance to oestradiol replacement therapy. Together, this review highlights the predominant modulatory, but nonetheless important, roles of ERβ in mediating the many effects of oestradiol upon adult brain function.
Collapse
Affiliation(s)
- R. J. Handa
- Department of Basic Medical Sciences, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA
| | - S. Ogawa
- Laboratory of Behavioral Neuroendocrinology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - J. M. Wang
- Department of Pathology, Pharmacology and Toxicology, Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
| | - A. E. Herbison
- Centre for Neuroendocrinology and Department of Physiology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
32
|
Keenan DM, Clarke IJ, Veldhuis JD. Noninvasive analytical estimation of endogenous GnRH drive: analysis using graded competitive GnRH-receptor antagonism and a calibrating pulse of exogenous GnRH. Endocrinology 2011; 152:4882-93. [PMID: 22028450 PMCID: PMC3230048 DOI: 10.1210/en.2011-1461] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Homeostatic control of endocrine systems proceeds via feedforward (agonistic, stimulatory) and feedback (antagonistic, inhibitory) interactions mediated via implicit dose-response functions. However, neither the feedback/feedforward pathways nor the dose-response interfaces are directly observed in vivo. Thus, the goal was to formulate and estimate an ensemble construct of time-varying feedback/feedforward interactions among GnRH, LH, and testosterone (T) in the male gonadal axis. The new analytical model revises and extends an earlier construct by: 1) allowing systemic T concentrations to inhibit hypothalamic GnRH output; 2) estimating GnRH outflow after injection of a calibrating pulse of biosynthetic GnRH; 3) framing the pituitary response to GnRH as a secretory burst, rather than continuous LH release; and 4) regressing feedback and feedforward ensemble parameters on age, rather than evaluating age dichotomously. Application of this methodology in 21 men aged 23-72 yr unveiled age-related 1) diminution of GnRH efficacy normalized for the decline in free T with age (P = 0.016), 2) potentiation of maximal T feedback onto (inhibition of) GnRH secretion (P = 0.006), and 3) accentuation of hypothalamic GnRH's sensitivity to T repression (P = 0.003). Outcomes were specific, because injected GnRH agonist and antagonist concentrations were invariant of age. We conclude that combining experimental and analytical strategies may provide a noninvasive means to investigate and decipher feedback determinants of unobserved endocrine signal(s).
Collapse
Affiliation(s)
- Daniel M Keenan
- Department of Statistics, University of Virginia, Charlottesville, Virginia 22904, USA
| | | | | |
Collapse
|
33
|
Shahjahan M, Hamabata T, Motohashi E, Doi H, Ando H. Differential expression of three types of gonadotropin-releasing hormone genes during the spawning season in grass puffer, Takifugu niphobles. Gen Comp Endocrinol 2010; 167:153-63. [PMID: 20138178 DOI: 10.1016/j.ygcen.2010.01.018] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Revised: 01/28/2010] [Accepted: 01/31/2010] [Indexed: 11/26/2022]
Abstract
Grass puffer, Takifugu niphobles, has unique spawning behavior; spawning occurs on beach only for several days around new moon and full moon from spring to early summer. To investigate the role of gonadotropin-releasing hormone (GnRH) in the reproductive function, genes encoding three types of GnRHs, namely seabream GnRH (sbGnRH), chicken GnRH-II (cGnRH-II) and salmon GnRH (sGnRH), were cloned and changes in their mRNA amounts were examined over the spawning season. In addition, changes in the pituitary gonadotropin subunit mRNAs and the plasma steroid hormones were examined over the spawning season. Fishes were assessed at four reproductive stages, i.e., in December (early maturation), in April (maturing), in May (spawning), and in July (post-spawning). Moreover, spawning fish just after releasing eggs and sperm were taken at a spawning bed. The amounts of sbGnRH mRNA were substantially elevated in May and the spawning fish in both sexes, concomitant with considerable elevations of follicle-stimulating hormone and luteinizing hormone beta subunit mRNAs and plasma estradiol-17beta (E(2)) and testosterone (T) levels. There were strong positive correlations between the sbGnRH mRNA and the plasma E(2) and T levels over the spawning season in both sexes. The amounts of cGnRH-II mRNA showed no noticeable changes except for an increase in the post-spawning females. The amounts of sGnRH mRNA in the males were significantly increased in May, but they were low in the spawning males. In the females, sGnRH mRNA increased from the maturing stage and reached a maximum in the post-spawning stage, in which a positive correlation with the plasma cortisol levels was observed. These specific changes suggest that the expression of three types of GnRH genes is differentially regulated during the spawning season, and sex steroids may be important for the differential expression of GnRH genes.
Collapse
Affiliation(s)
- Md Shahjahan
- Laboratory of Advanced Animal and Marine Bioresources, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka 812-8581, Japan
| | | | | | | | | |
Collapse
|
34
|
Kurian JR, Olesen KM, Auger AP. Sex differences in epigenetic regulation of the estrogen receptor-alpha promoter within the developing preoptic area. Endocrinology 2010; 151:2297-305. [PMID: 20237133 PMCID: PMC2869250 DOI: 10.1210/en.2009-0649] [Citation(s) in RCA: 157] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Sex differences in the brain are largely organized by a testicular hormone surge that occurs in males shortly after birth. Although this hormone surge is transient, sex differences in brain and behavior are lasting. Here we describe a sex difference in DNA methylation of the estrogen receptor-alpha (ERalpha) promoter region within the developing rat preoptic area, with males exhibiting more DNA methylation within the ERalpha promoter than females. More importantly, we report that simulating maternal grooming, a form of maternal interaction that is sexually dimorphic with males experiencing more than females during the neonatal period, effectively masculinizes female ERalpha promoter methylation and gene expression. This suggests natural variations in maternal care that are directed differentially at males vs. females can influence sex differences in the brain by creating sexually dimorphic DNA methylation patterns. We also find that the early estradiol exposure may contribute to sex differences in DNA methylation patterns. This suggests that early social interaction and estradiol exposure may converge at the genome to organize lasting sex differences in the brain via epigenetic differentiation.
Collapse
Affiliation(s)
- Joseph R Kurian
- Psychology Department, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | | | | |
Collapse
|
35
|
Park SW, Huang WH, Persaud SD, Wei LN. RIP140 in thyroid hormone-repression and chromatin remodeling of Crabp1 gene during adipocyte differentiation. Nucleic Acids Res 2010; 37:7085-94. [PMID: 19778926 PMCID: PMC2790899 DOI: 10.1093/nar/gkp780] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Cellular retinoic acid binding protein 1 (Crabp1) gene is biphasically (proliferation versus differentiation) regulated by thyroid hormone (T3) in 3T3-L1 cells. This study examines T3-repression of Crabp1 gene during adipocyte differentiation. T3 repression of Crabp1 requires receptor interacting protein 140 (RIP140). During differentiation, the juxtaposed chromatin configuration of Crabp1 promoter with its upstream region is maintained, but the 6-nucleosomes spanning thyroid hormone response element to transcription initiation site slide bi-directionally, with the third nucleosome remaining at the same position throughout differentiation. On the basal promoter, RIP140 replaces coactivators GRIP1 and PCAF and forms a repressive complex with CtBP1, HDAC3 and G9a. Initially active chromatin marks on this promoter, histone modifications H3-Ac and H3K4-me3, are weakened whereas repressive chromatin marks, H3K9-me3 and H3K27-me3 modification and recruitment of G9a, HP1α, HP1γ and H1, are intensified. This is the first study to examine chromatin remodeling, during the phase of hormone repression, of a bi-directionally regulated hormone target gene, and provides evidence for a functional role of RIP140 in chromatin remodeling to repress hormone target gene expression.
Collapse
Affiliation(s)
- Sung Wook Park
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | | | | | | |
Collapse
|
36
|
Novaira HJ, Ng Y, Wolfe A, Radovick S. Kisspeptin increases GnRH mRNA expression and secretion in GnRH secreting neuronal cell lines. Mol Cell Endocrinol 2009; 311:126-34. [PMID: 19576263 PMCID: PMC3534746 DOI: 10.1016/j.mce.2009.06.011] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2009] [Revised: 06/02/2009] [Accepted: 06/19/2009] [Indexed: 12/27/2022]
Abstract
Kisspeptins, and their G-protein coupled receptor 54 (GPR54), are key components in the regulation of gonadotropin-releasing hormone (GnRH) secretion in humans and other mammals. Several studies demonstrate that the central or systemic administration of kisspeptin increases GnRH and gonadotropin secretion in both prepubertal and adult animals; however, the cellular targets and intracellular mechanisms of action in the central reproductive axis are unclear. In this study, we documented the presence of GPR54 in two GnRH secreting neuronal cell lines (GT1-7 and GN11). Kisspeptin treatment increases GnRH secretion and GnRH mRNA levels in a dose and time dependent manner. 10(-9)M kisspeptin maximally stimulated GnRH secretion by 2-fold and GnRH mRNA levels up to 4-fold after 4h of treatment in both cell lines. Negative regulation by 17beta-estradiol of GnRH secretion and GnRH mRNA was antagonized by kisspeptin. Co-treatment with kisspeptin and 17beta-estradiol increased GnRH secretion by 2-fold and GnRH mRNA by 4-fold over estradiol alone in both cell lines. Intracellular signaling pathway studies showed that an ERK1/2 MAPK inhibitor (PD98059) and a PI3K inhibitor, LY29402, attenuated the effects of kisspeptin on GnRH mRNA modulation. Furthermore, Western blot analysis showed that phosphorylation of both MAPK and Akt substrates increased with kisspeptin treatment. This work demonstrates that the kisspeptin-GPR54 system plays a significant role stimulating GnRH secretion and positive regulation of GnRH mRNA levels in GnRH neurons in culture, and also, demonstrates the activation of MAPK and Akt signaling pathways by kisspeptin in GT1-7 and GN11 cell lines.
Collapse
Affiliation(s)
- Horacio J Novaira
- Department of Pediatrics, Division of Endocrinology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| | | | | | | |
Collapse
|