1
|
Hackwell E, Ladyman SR, Clarkson J, McQullian HJ, Boehm U, Herbison AE, Brown R, Grattan DR. Prolactin-mediates a lactation-induced suppression of arcuate kisspeptin neuronal activity necessary for lactational infertility in mice. eLife 2025; 13:RP94570. [PMID: 39819370 PMCID: PMC11741520 DOI: 10.7554/elife.94570] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025] Open
Abstract
The specific role that prolactin plays in lactational infertility, as distinct from other suckling or metabolic cues, remains unresolved. Here, deletion of the prolactin receptor (Prlr) from forebrain neurons or arcuate kisspeptin neurons resulted in failure to maintain normal lactation-induced suppression of estrous cycles. Kisspeptin immunoreactivity and pulsatile LH secretion were increased in these mice, even in the presence of ongoing suckling stimulation and lactation. GCaMP fibre photometry of arcuate kisspeptin neurons revealed that the normal episodic activity of these neurons is rapidly suppressed in pregnancy and this was maintained throughout early lactation. Deletion of Prlr from arcuate kisspeptin neurons resulted in early reactivation of episodic activity of kisspeptin neurons prior to a premature return of reproductive cycles in early lactation. These observations show dynamic variation in arcuate kisspeptin neuronal activity associated with the hormonal changes of pregnancy and lactation, and provide direct evidence that prolactin action on arcuate kisspeptin neurons is necessary for suppressing fertility during lactation in mice.
Collapse
Affiliation(s)
- Eleni Hackwell
- Centre for Neuroendocrinology, Dunedin, New Zealand
- Department of Anatomy, Dunedin, New Zealand
| | - Sharon R Ladyman
- Centre for Neuroendocrinology, Dunedin, New Zealand
- Department of Anatomy, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Jenny Clarkson
- Centre for Neuroendocrinology, Dunedin, New Zealand
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - H James McQullian
- Centre for Neuroendocrinology, Dunedin, New Zealand
- Department of Anatomy, Dunedin, New Zealand
| | - Ulrich Boehm
- Saarland University School of Medicine, Centre for Molecular Signalling (PZMS), Experimental Pharmacology, Homburg, Germany
| | - Allan Edward Herbison
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Rosemary Brown
- Centre for Neuroendocrinology, Dunedin, New Zealand
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - David R Grattan
- Centre for Neuroendocrinology, Dunedin, New Zealand
- Department of Anatomy, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| |
Collapse
|
2
|
Watanabe Y, Fisher L, Campbell RE, Jasoni CL. Developmental expression patterns of gonadal hormone receptors in arcuate kisspeptin and GABA neurons of the postnatal female mouse. J Neuroendocrinol 2025; 37:e13477. [PMID: 39605295 DOI: 10.1111/jne.13477] [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: 07/18/2024] [Revised: 10/29/2024] [Accepted: 11/17/2024] [Indexed: 11/29/2024]
Abstract
The arcuate nucleus of the hypothalamus (ARC) is central in the neuronal regulation of fertility and reproduction through translating gonadal steroid hormone cues into the GnRH signaling pathway in the brain. Evidence suggests that circulating gonadal steroids play an important role in modulating female reproduction via kisspeptin and γ-aminobutyric acid (GABA) neurons in the ARC in both development and adulthood. However, the temporal onset of these ARC neurons' sensitivity to gonadal steroids is unknown. Using RNAscope® in situ hybridization, we localized androgen receptor (Ar), estrogen receptor alpha (Esr1), and progesterone receptor (Pgr) expression in ARC kisspeptin or GABA neurons of female mice at postnatal day (P)4, P8, P12, P20, and P60. A probe that binds to kiss1 mRNA or vGat mRNA was used to produce signal in kisspeptin or GABA neurons, respectively. In adult, we identified that the vast majority of kisspeptin neurons coexpressed Esr1 (95%) and Pgr (93%), while a smaller proportion coexpressed Ar (66%). Similar proportions of Ar- or Esr1-positive kisspeptin neurons were seen from P4, suggesting that kisspeptin neurons develop adult-like sensitivity to androgen and estrogen in early postnatal life. In contrast, the proportion of Pgr-positive kisspeptin cells in early life was significantly lower than in adulthood, suggesting that progesterone sensitivity develops over time in the ARC kisspeptin population. ARC GABA neurons also colocalized with Ar (70%), Esr1 (64%), or Pgr (85%) in adulthood. GABA neurons continuously expressed Esr1 or Pgr from the postnatal stages to adulthood, while the proportion of Ar-positive GABA neurons gradually increased from P4 (24%) to P20 (59%). These results suggest that while ARC GABA neurons can respond to circulating estrogen and progesterone from early postnatal ages, this same population may become more sensitive to androgens during later postnatal life. Our findings identified the expression patterns of Ar, Esr1, and Pgr by ARC kisspeptin and GABA neurons during early postnatal life. These data provide the understanding for the hormone sensitivity of these populations during early postnatal life, the critical time for the formation and regulation of female reproductive physiology.Esr1 (95%) and Pgr (93%), while a smaller proportion coexpressed Ar (66%). Similar proportions of Ar- or Esr1-positive kisspeptin neurons were seen from P4, suggesting that kisspeptin neurons develop adult-like sensitivity to androgen and estrogen in early postnatal life. In contrast, the proportion of Pgr-positive kisspeptin cells in early life was significantly lower than in adulthood, suggesting that progesterone sensitivity develops over time in the ARC kisspeptin population. ARC GABA neurons also colocalized with Ar (70%), Esr1 (64%), or Pgr (85%) in adulthood. GABA neurons continuously expressed Esr1 or Pgr from the postnatal stages to adulthood, while the proportion of Ar-positive GABA neurons gradually increased from P4 (24%) to P20 (59%). These results suggest that while ARC GABA neurons can respond to circulating estrogen and progesterone from early postnatal ages, this same population may become more sensitive to androgens during later postnatal life. Our findings identified the expression patterns of Ar, Esr1, and Pgr by ARC kisspeptin and GABA neurons during early postnatal life. These data provide the understanding for the hormone sensitivity of these populations during early postnatal life, the critical time for the formation and regulation of female reproductive physiology.
Collapse
Affiliation(s)
- Yugo Watanabe
- Centre for Neuroendocrinology, Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
- Department of Biochemistry, School of Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Lorryn Fisher
- Centre for Neuroendocrinology, Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Rebecca E Campbell
- Centre for Neuroendocrinology, Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Christine L Jasoni
- Centre for Neuroendocrinology, Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, New Zealand
| |
Collapse
|
3
|
González-Flores O, Garcia-Juárez M, Tecamachaltzi-Silvarán MB, Lucio RA, Ordoñez RD, Pfaus JG. Cellular and molecular mechanisms of action of ovarian steroid hormones. I: Regulation of central nervous system function. Neurosci Biobehav Rev 2024; 167:105937. [PMID: 39510217 DOI: 10.1016/j.neubiorev.2024.105937] [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: 08/27/2024] [Revised: 10/25/2024] [Accepted: 10/31/2024] [Indexed: 11/15/2024]
Abstract
The conventional way steroid hormones work through receptors inside cells is widely acknowledged. There are unanswered questions about what happens to the hormone in the end and why there isn't always a strong connection between how much tissue takes up and its biological effects through receptor binding. Steroid hormones can also have non-traditional effects that happen quickly but don't involve entering the cell. Several possible mechanisms for these non-traditional actions include (a) changes in membrane fluidity, (b) steroid hormones acting on receptors on the outer surface of cells, (c) steroid hormones regulating GABAA receptors on cell membranes, and (d) activation of steroid receptors by factors like EGF, IGF-1, and dopamine. Data also suggests that steroid hormones may be inserted into DNA through receptors, acting as transcription factors. These proposed new mechanisms of action should not be seen as challenging the conventional mechanism. Instead, they contribute to a more comprehensive understanding of how hormones work, allowing for rapid, short-term, and prolonged effects to meet the body's physiological needs.
Collapse
Affiliation(s)
- Oscar González-Flores
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, Tlaxcala, Mexico.
| | - Marcos Garcia-Juárez
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, Tlaxcala, Mexico
| | | | - Rosa Angélica Lucio
- Centro Tlaxcala de Biología de la Conducta, Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico
| | - Raymundo Domínguez Ordoñez
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, Tlaxcala, Mexico; Licenciatura en Ingeniería Agronómica y Zootecnia, Complejo Regional Centro, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - James G Pfaus
- Center for Sexual Health and Intervention, Czech National Institute of Mental Health, Klecany, Czech Republic; Department of Psychology and Life Sciences, Faculty of Humanities, Charles University, Prague, Czech Republic
| |
Collapse
|
4
|
Cruz M, Howles CM. Clinical outcomes of three follitropin alfa preparations for ovarian stimulation using an oral micronized progesterone-primed protocol in an oocyte donation program. Front Endocrinol (Lausanne) 2024; 15:1451668. [PMID: 39398338 PMCID: PMC11467753 DOI: 10.3389/fendo.2024.1451668] [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: 06/19/2024] [Accepted: 09/09/2024] [Indexed: 10/15/2024] Open
Abstract
Introduction This large multicenter study aimed to evaluate clinical outcomes using three follitropin alfa preparations within a progestin-primed ovarian stimulation (PPOS) protocol, while identifying contributing factors to cycle success. Methods A retrospective, anonymized cohort analysis was conducted on donor-recipient cycles from 12 clinics during 2019 to 2021. 7389 oocyte donors underwent ovarian stimulation (OS) with three follitropin alfa preparations (Ovaleap® [n=3231], Bemfola® [n=3542], Gonal-F® [n=616]) were included. Stimulation began on cycle days 2 or 3 with daily administration of 150-225 IU follitropin alfa. 10 mg medroxyprogesterone acetate (MPA) was administered daily until GnRH agonist trigger using a single dose of 0.2mg GnRH agonist for final follicular maturation. Statistical analysis included ANOVA, Chi-squared, and logistic regression. Results Whilst there were some differences in patient and stimulation characteristics, including donor age and number of retrieved oocytes, clinical variables did not significantly differ among the three study groups. Linear regression revealed donor age [0.986 (0.974-0.999)] and number of mature oocytes [1.027 (1.007-1.047)] significantly impacted ongoing pregnancy rates, while the type of follitropin alfa [1.048 (0.956-1.149)] used did not. No significant differences were observed in the cumulative live birth rate (CLBR) among oocytes obtained from stimulation with Bemfola (64.9%), Gonal-F (64.1%) and Ovaleap (66.1%), p= 0.385. Discussion This study demonstrated comparable clinical outcomes and CLBR between biosimilars and the reference product of follitropin alfa within PPOS protocols, hence they are interchangeable in a real-world patient setting.
Collapse
Affiliation(s)
- María Cruz
- IVI-RMA Global Headquarters, Madrid, Spain
| | - Colin M. Howles
- ARIES Consulting Sàrl, Geneva, Switzerland
- Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| |
Collapse
|
5
|
Dillon KM, Lohr DB, Novak AG, Petriv AMV, Neifert NT, Moore AM. Deletion of Nuclear Progesterone Receptors From Kisspeptin Cells Does Not Impair Negative Feedback in Female Mice. Endocrinology 2024; 165:bqae121. [PMID: 39253941 DOI: 10.1210/endocr/bqae121] [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: 07/10/2024] [Revised: 09/03/2024] [Accepted: 09/08/2024] [Indexed: 09/11/2024]
Abstract
Reproductive function in mammals depends on the ability of progesterone (P4) to suppress pulsatile gonadotrophin-releasing hormone (GnRH) and luteinizing hormone (LH) secretion in a homeostatic-negative feedback loop. Previous research identified that cells upstream from GnRH neurons expressing the nuclear progesterone receptor (PGR) are required for P4-negative feedback. However, the identity of these cells and the mechanism by which they reduce GnRH/LH pulsatile secretion is unknown. We aimed to address the hypothesis that PGR expressed by a neural population in the arcuate nucleus recently identified as the GnRH pulse generator, cells expressing kisspeptin, neurokinin B, and dynorphin (KNDy cells), mediate P4-negative feedback. To achieve this, we used female mice with the PGR gene conditionally deleted from kisspeptin cells (KPRKO mice) and observed a substantial decrease in the percentage of KNDy neurons coexpressing PGR messenger RNA (mRNA) (11% in KPRKO mice vs 86% in wild-type [WT] mice). However, KPRKO mice did not display changes in the frequency or amplitude of LH pulses in diestrus or estrus, nor in the ability of exogenous P4 to blunt a postcastration increase in LH. Further, mRNA expression of arcuate kisspeptin and dynorphin, which are excitatory and inhibitory to GnRH secretion, respectively, remained unaltered in KPRKO mice compared to WT controls. Together, these findings show that the near-complete loss of PGR signaling from KNDy cells does not affect negative feedback regulation of GnRH pulse generation in mice, suggesting that feedback through this receptor can occur via a small number of KNDy cells or a yet unidentified cell population.
Collapse
Affiliation(s)
- Kendra M Dillon
- Department of Biological Sciences, Brain Health Research Institute, Kent State University, Kent, OH 44242-0001, USA
| | - Dayanara B Lohr
- Department of Biological Sciences, Brain Health Research Institute, Kent State University, Kent, OH 44242-0001, USA
| | - Alyssa G Novak
- Department of Biological Sciences, Brain Health Research Institute, Kent State University, Kent, OH 44242-0001, USA
| | - Anna-Maria V Petriv
- Department of Biological Sciences, Brain Health Research Institute, Kent State University, Kent, OH 44242-0001, USA
| | - Nicole T Neifert
- Department of Biological Sciences, Brain Health Research Institute, Kent State University, Kent, OH 44242-0001, USA
| | - Aleisha M Moore
- Department of Biological Sciences, Brain Health Research Institute, Kent State University, Kent, OH 44242-0001, USA
| |
Collapse
|
6
|
Dardente H, Lomet D, Robert V, Lasserre O, Gonzalez AA, Mialhe X, Beltramo M. Photoperiod, but not progesterone, has a strong impact upon the transcriptome of the medio-basal hypothalamus in female goats and ewes. Mol Cell Endocrinol 2024; 588:112216. [PMID: 38556161 DOI: 10.1016/j.mce.2024.112216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 03/11/2024] [Accepted: 03/24/2024] [Indexed: 04/02/2024]
Abstract
Photoperiod is the main environmental driver of seasonal responses in organisms living at temperate and polar latitudes. Other external cues such as food and temperature, and internal cues including hormones, intervene to fine-tune phasing of physiological functions to the solar year. In mammals, the medio-basal hypothalamus (MBH) is the key integrator of these cues, which orchestrates a wide array of seasonal functions, including breeding. Here, using RNAseq and RT-qPCR, we demonstrate that molecular components of the photoperiodic response previously identified in ewes are broadly conserved in does (female goats, Capra hircus), with a common core of ∼50 genes. This core group can be defined as the "MBH seasonal trancriptome", which includes key players of the pars tuberalis-tanycytes neuroendocrine retrograde pathway that governs intra-MBH photoperiodic switches of triiodothyronine (T3) production (Tshb, Eya3, Dio2 and SlcO1c1), the two histone methyltransferases Suv39H2 and Ezh2 and the secreted protein Vmo1. Prior data in ewes revealed that T3 and estradiol (E2), both key hormones for the proper timing of seasonal breeding, differentially impact the MBH seasonal transcriptome, and identified cellular and molecular targets through which these hormones might act. In contrast, information regarding the potential impact of progesterone (P4) upon the MBH transcriptome was nonexistent. Here, we demonstrate that P4 has no discernible transcriptional impact in either does or ewes. Taken together, our data show that does and ewes possess a common core set of photoperiod-responsive genes in the MBH and conclusively demonstrate that P4 is not a key regulator of the MBH transcriptome.
Collapse
Affiliation(s)
- Hugues Dardente
- INRAE, CNRS, Université de Tours, PRC, 37380, Nouzilly, France.
| | - Didier Lomet
- INRAE, CNRS, Université de Tours, PRC, 37380, Nouzilly, France
| | - Vincent Robert
- INRAE, CNRS, Université de Tours, PRC, 37380, Nouzilly, France
| | | | - Anne-Alicia Gonzalez
- MGX-Montpellier GenomiX, Univ. Montpellier, CNRS, INSERM, 34094, Montpellier, France
| | - Xavier Mialhe
- MGX-Montpellier GenomiX, Univ. Montpellier, CNRS, INSERM, 34094, Montpellier, France
| | | |
Collapse
|
7
|
Vidal MDM, Martínez F, Rodríguez I, Polyzos NP. Ovarian response and embryo ploidy following oral micronized progesterone-primed ovarian stimulation versus GnRH antagonist protocol. A prospective study with repeated ovarian stimulation cycles. Hum Reprod 2024; 39:1098-1104. [PMID: 38498835 DOI: 10.1093/humrep/deae047] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 01/21/2024] [Indexed: 03/20/2024] Open
Abstract
STUDY QUESTION Is there any difference in ovarian response and embryo ploidy following progesterone-primed ovarian stimulation (PPOS) using micronized progesterone or GnRH antagonist protocol? SUMMARY ANSWER Pituitary downregulation with micronized progesterone as PPOS results in higher number of oocytes retrieved and a comparable number of euploid blastocysts to a GnRH antagonist protocol. WHAT IS KNOWN ALREADY Although the GnRH antagonist is considered by most the gold standard protocol for controlling the LH surge during ovarian stimulation (OS) for IVF/ICSI, PPOS protocols are being increasingly used in freeze-all protocols. Still, despite the promising results of PPOS protocols, an early randomized trial reported potentially lower live births in recipients of oocytes resulting following downregulation with medroxyprogesterone acetate as compared with a GnRH antagonist protocol. The scope of the current prospective study was to investigate whether PPOS with micronized progesterone results in an equivalent yield of euploid blastocysts to a GnRH antagonist protocol. STUDY DESIGN, SIZE, DURATION In this prospective study, performed between September 2019 to January 2022, 44 women underwent two consecutive OS protocols within a period of 6 months in a GnRH antagonist protocol or in a PPOS protocol with oral micronized progesterone. PARTICIPANTS/MATERIALS, SETTING, METHODS Overall, 44 women underwent two OS cycles with an identical fixed dose of rFSH (225 or 300 IU) in both cycles. Downregulation in the first cycles was performed with the use of a flexible GnRH antagonist protocol (0.25 mg per day as soon as one follicle of 14 mm) and consecutively, after a washout period of 1 month, control of LH surge was performed with 200 mg of oral micronized progesterone from stimulation Day 1. After the completion of both cycles, all generated blastocysts underwent genetic analysis for aneuploidy screening (preimplantation genetic testing for aneuplody, PGT-A). MAIN RESULTS AND THE ROLE OF CHANCE Comparisons between protocols did not reveal differences between the duration of OS. The hormonal profile on the day of trigger revealed statistically significant differences between protocols in all the tested hormones except for FSH: with significantly higher serum E2 levels, more elevated LH levels and higher progesterone levels in PPOS cycles as compared with antagonist cycles, respectively. Compared with the GnRH antagonist protocol, the PPOS protocol resulted in a significantly higher number of oocytes (12.7 ± 8.09 versus 10.3 ± 5.84; difference between means [DBM] -2.4 [95% CI -4.1 to -0.73]), metaphase II (9.1 ± 6.12 versus 7.3 ± 4.15; DBM -1.8 [95% CI -3.1 to -0.43]), and 2 pronuclei (7.1 ± 4.99 versus 5.7 ± 3.35; DBM -1.5 [95% CI -2.6.1 to -0.32]), respectively. Nevertheless, no differences were observed regarding the mean number of blastocysts between the PPOS and GnRH antagonist protocols (2.9 ± 2.11 versus 2.8 ± 2.12; DBM -0.07 [95% CI -0.67 to 0.53]) and the mean number of biopsied blastocysts (2.9 ± 2.16 versus 2.9 ± 2.15; DBM -0.07 [95% CI -0.70 to 0.56]), respectively. Concerning the euploidy rates per biopsied embryo, a 29% [95% CI 21.8-38.1%] and a 35% [95% CI 26.6-43.9%] were noticed in the PPOS and antagonist groups, respectively. Finally, no difference was observed for the primary outcome, with a mean number of euploid embryos of 0.86 ± 0.90 versus 1.00 ± 1.12 for the comparison of PPOS versus GnRh antagonist. LIMITATIONS, REASONS FOR CAUTION The study was powered to detect differences in the mean number of euploid embryos and not in terms of pregnancy outcomes. Additionally, per protocol, there was no randomization, the first cycle was always a GnRH antagonist cycle and the second a PPOS with 1 month of washout period in between. WIDER IMPLICATIONS OF THE FINDINGS In case of a freeze-all protocol, clinicians may safely consider oral micronized progesterone to control the LH surge and patients could benefit from the advantages of a medication of oral administration, with a potentially higher number of oocytes retrieved at a lower cost, without any compromise in embryo ploidy rates. STUDY FUNDING/COMPETING INTEREST(S) This research was supported by an unrestricted grant from Theramex. N.P.P. has received Research grants from Merck Serono, Organon, Ferring Pharmaceutical, Roche, Theramex, IBSA, Gedeon Richter, and Besins Healthcare; honoraria for lectures from: Merck Serono, Organon, Ferring Pharmaceuticals, Besins International, Roche Diagnostics, IBSA, Theramex, and Gedeon Richter; consulting fees from Merck Serono, Organon, Besins Healthcare, and IBSA. M.d.M.V., F.M., and I.R. declared no conflicts of interest. TRIAL REGISTRATION NUMBER The study was registered at Clinical Trials Gov. (NCT04108039).
Collapse
Affiliation(s)
- M Del Mar Vidal
- Dexeus Fertility , Hospital Universitari Dexeus, Barcelona 08014, Spain
| | | | - Ignacio Rodríguez
- Dexeus Fertility , Hospital Universitari Dexeus, Barcelona 08014, Spain
| | | |
Collapse
|
8
|
Orda MA, Fowler PMPT, Tayo LL. Modular Hub Genes in DNA Microarray Suggest Potential Signaling Pathway Interconnectivity in Various Glioma Grades. BIOLOGY 2024; 13:206. [PMID: 38666818 PMCID: PMC11048586 DOI: 10.3390/biology13040206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/07/2024] [Accepted: 03/21/2024] [Indexed: 04/28/2024]
Abstract
Gliomas have displayed significant challenges in oncology due to their high degree of invasiveness, recurrence, and resistance to treatment strategies. In this work, the key hub genes mainly associated with different grades of glioma, which were represented by pilocytic astrocytoma (PA), oligodendroglioma (OG), anaplastic astrocytoma (AA), and glioblastoma multiforme (GBM), were identified through weighted gene co-expression network analysis (WGCNA) of microarray datasets retrieved from the Gene Expression Omnibus (GEO) database. Through this, four highly correlated modules were observed to be present across the PA (GSE50161), OG (GSE4290), AA (GSE43378), and GBM (GSE36245) datasets. The functional annotation and pathway enrichment analysis done through the Database for Annotation, Visualization, and Integrated Discovery (DAVID) showed that the modules and hub genes identified were mainly involved in signal transduction, transcription regulation, and protein binding, which collectively deregulate several signaling pathways, mainly PI3K/Akt and metabolic pathways. The involvement of several hub genes primarily linked to other signaling pathways, including the cAMP, MAPK/ERK, Wnt/β-catenin, and calcium signaling pathways, indicates potential interconnectivity and influence on the PI3K/Akt pathway and, subsequently, glioma severity. The Drug Repurposing Encyclopedia (DRE) was used to screen for potential drugs based on the up- and downregulated hub genes, wherein the synthetic progestin hormones norgestimate and ethisterone were the top drug candidates. This shows the potential neuroprotective effect of progesterone against glioma due to its influence on EGFR expression and other signaling pathways. Aside from these, several experimental and approved drug candidates were also identified, which include an adrenergic receptor antagonist, a PPAR-γ receptor agonist, a CDK inhibitor, a sodium channel blocker, a bradykinin receptor antagonist, and a dopamine receptor agonist, which further highlights the gene network as a potential therapeutic avenue for glioma.
Collapse
Affiliation(s)
- Marco A. Orda
- School of Chemical, Biological, and Materials Engineering and Sciences, Mapúa University, Manila City 1002, Philippines; (M.A.O.); (P.M.P.T.F.)
- School of Graduate Studies, Mapúa University, Manila City 1002, Philippines
| | - Peter Matthew Paul T. Fowler
- School of Chemical, Biological, and Materials Engineering and Sciences, Mapúa University, Manila City 1002, Philippines; (M.A.O.); (P.M.P.T.F.)
- Department of Biology, School of Health Sciences, Mapúa University, Makati City 1203, Philippines
| | - Lemmuel L. Tayo
- School of Chemical, Biological, and Materials Engineering and Sciences, Mapúa University, Manila City 1002, Philippines; (M.A.O.); (P.M.P.T.F.)
- Department of Biology, School of Health Sciences, Mapúa University, Makati City 1203, Philippines
| |
Collapse
|
9
|
Krishnakumar S, Malavika RN, Nair SV, Menon D, Paul-Prasanth B. Nano-graphene oxide particles induce inheritable anomalies through altered gene expressions involved in oocyte maturation. Nanotoxicology 2024; 18:160-180. [PMID: 38449436 DOI: 10.1080/17435390.2024.2325615] [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: 01/29/2024] [Accepted: 02/26/2024] [Indexed: 03/08/2024]
Abstract
The inheritable impact of exposure to graphene oxide nanoparticles (GO NPs) on vertebrate germline during critical windows of gamete development remain undetermined to date. Here, we analyzed the transgenerational effects of exposure to nano-graphene oxide particles (nGO) synthesized in house with lateral dimensions 300-600 nm and surface charge of -36.8 mV on different developmental stages of germ cells (GCs): (1) during GCs undergoing early development and differentiation, and (2) during GCs undergoing gametogenesis and maturation in adulthood. Biocompatibility analyses in Japanese medaka embryos showed lethality above 1 µg/ml and also an aberrant increase in germ cell count of both males and females at doses below the lethal dose. However, no lethality or anomalies were evident in adults up to 45 µg/ml. Long term exposure of embryos and adults for 21 days resulted in reduced fecundity. This effect was transmitted to subsequent generations, F1 and F2. Importantly, the inheritable effects of nGO in adults were pronounced at a high dose of 10 µg/ml, while 1 µg/ml showed no impact on the germline indicating lower doses used in this study to be safe. Further, expressions of selected genes that adversely affected oocyte maturation were enhanced in F1 and F2 individuals. Interestingly, the inheritance patterns differed corresponding to the stage at which the fish received the exposure.
Collapse
Affiliation(s)
- Sreelakshmi Krishnakumar
- School of Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi Campus, Kerala, India
| | - Raghunath Nair Malavika
- School of Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi Campus, Kerala, India
| | - Shantikumar V Nair
- School of Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi Campus, Kerala, India
| | - Deepthy Menon
- School of Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi Campus, Kerala, India
| | - Bindhu Paul-Prasanth
- School of Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi Campus, Kerala, India
| |
Collapse
|
10
|
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
|
11
|
Liu M, Zhang J, Zhou Y, Xiong S, Zhou M, Wu L, Liu Q, Chen Z, Jiang H, Yang J, Liu Y, Wang Y, Chen C, Huang L. Gut microbiota affects the estrus return of sows by regulating the metabolism of sex steroid hormones. J Anim Sci Biotechnol 2023; 14:155. [PMID: 38115159 PMCID: PMC10731813 DOI: 10.1186/s40104-023-00959-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 11/06/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND Sex hormones play important roles in the estrus return of post-weaning sows. Previous studies have demonstrated a complex and bi-directional regulation between sex hormones and gut microbiota. However, the extent to which the gut microbiota affects estrus return of post-weaning sows is largely unknown. RESULTS In this study, we first screened 207 fecal samples from well-phenotyped sows by 16S rRNA gene sequencing and identified significant associations between microbes and estrus return of post-weaning sows. Using metagenomic sequencing data from 85 fecal samples, we identified 37 bacterial species that were significantly associated with estrus return. Normally returning sows were characterized by increased abundances of L. reuteri and P. copri and decreased abundances of B. fragilis, S. suis, and B. pseudolongum. The changes in gut microbial composition significantly altered the functional capacity of steroid hormone biosynthesis in the gut microbiome. The results were confirmed in a validation cohort. Significant changes in sex steroid hormones and related compounds were found between normal and non-return sows via metabolome analysis. An integrated analysis of differential bacterial species, metagenome, and fecal metabolome provided evidence that normal return-associated bacterial species L. reuteri and Prevotella spp. participated in the degradation of pregnenolone, progesterone, and testosterone, thereby promoting estrogen biosynthesis. Furthermore, the microbial metabolites related to sow energy and nutrient supply or metabolic disorders also showed relationships with sow estrus return. CONCLUSIONS An integrated analysis of differentially abundant bacterial species, metagenome, and fecal metabolome revealed the involvement of L. reuteri and Prevotella spp. in sow estrus return. These findings provide deep insight into the role of gut microbiota in the estrus return of post-weaning sows and the complex cross-talk between gut microbiota and sex hormones, suggesting that the manipulation of the gut microbiota could be an effective strategy to improve sow estrus return after weaning.
Collapse
Affiliation(s)
- Min Liu
- National Key Laboratory of Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Jia Zhang
- National Key Laboratory of Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Yunyan Zhou
- National Key Laboratory of Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Shuqi Xiong
- National Key Laboratory of Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Mengqing Zhou
- National Key Laboratory of Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Lin Wu
- National Key Laboratory of Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Qin Liu
- National Key Laboratory of Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Zhe Chen
- National Key Laboratory of Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Hui Jiang
- National Key Laboratory of Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Jiawen Yang
- National Key Laboratory of Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Yuxin Liu
- National Key Laboratory of Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Yaxiang Wang
- National Key Laboratory of Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Congying Chen
- National Key Laboratory of Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China.
| | - Lusheng Huang
- National Key Laboratory of Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China.
| |
Collapse
|
12
|
Liu ZB, Zhang JB, Li SP, Yu WJ, Pei N, Jia HT, Li Z, Lv WF, Wang J, Kim NH, Yuan B, Jiang H. ID3 regulates progesterone synthesis in bovine cumulus cells through modulation of mitochondrial function. Theriogenology 2023; 209:141-150. [PMID: 37393744 DOI: 10.1016/j.theriogenology.2023.06.035] [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: 04/06/2023] [Revised: 06/05/2023] [Accepted: 06/23/2023] [Indexed: 07/04/2023]
Abstract
DNA binding inhibitory factor 3 (ID3) has been shown to have a key role in maintaining proliferation and differentiation. It has been suggested that ID3 may also affect mammalian ovarian function. However, the specific roles and mechanisms are unclear. In this study, the expression level of ID3 in cumulus cells (CCs) was inhibited by siRNA, and the downstream regulatory network of ID3 was uncovered by high-throughput sequencing. The effects of ID3 inhibition on mitochondrial function, progesterone synthesis, and oocyte maturation were further explored. The GO and KEGG analysis results showed that after ID3 inhibition, differentially expressed genes, including StAR, CYP11A1, and HSD3B1, were involved in cholesterol-related processes and progesterone-mediated oocyte maturation. Apoptosis in CC was increased, while the phosphorylation level of ERK1/2 was inhibited. During this process, mitochondrial dynamics and function were disrupted. In addition, the first polar body extrusion rate, ATP production and antioxidation capacity were reduced, which suggested that ID3 inhibition led to poor oocyte maturation and quality. The results will provide a new basis for understanding the biological roles of ID3 as well as cumulus cells.
Collapse
Affiliation(s)
- Zi-Bin Liu
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, Jilin, 130062, People's Republic of China
| | - Jia-Bao Zhang
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, Jilin, 130062, People's Republic of China
| | - Sheng-Peng Li
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, Jilin, 130062, People's Republic of China
| | - Wen-Jie Yu
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, Jilin, 130062, People's Republic of China
| | - Na Pei
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, Jilin, 130062, People's Republic of China
| | - Hai-Tao Jia
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, Jilin, 130062, People's Republic of China
| | - Ze Li
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, Jilin, 130062, People's Republic of China
| | - Wen-Fa Lv
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin, 130118, People's Republic of China
| | - Jun Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin, 130118, People's Republic of China
| | - Nam-Hyung Kim
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, Jilin, 130062, People's Republic of China; Department of Animal Science, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Bao Yuan
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, Jilin, 130062, People's Republic of China.
| | - Hao Jiang
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, Jilin, 130062, People's Republic of China.
| |
Collapse
|
13
|
Ruddenklau A, Glendining K, Prescott M, Campbell RE. Validation of a new Custom Polyclonal Progesterone Receptor Antibody for Immunohistochemistry in the Female Mouse Brain. J Endocr Soc 2023; 7:bvad113. [PMID: 37693686 PMCID: PMC10492226 DOI: 10.1210/jendso/bvad113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Indexed: 09/12/2023] Open
Abstract
Immunohistochemical visualization of progesterone receptor (PR)-expressing cells in the brain is a powerful technique to investigate the role of progesterone in the neuroendocrine regulation of fertility. A major obstacle to the immunohistochemical visualization of progesterone-sensitive cells in the rodent brain has been the discontinuation of the commercially produced A0098 rabbit polyclonal PR antibody by DAKO. To address the unavailability of this widely used PR antibody, we optimized and evaluated 4 alternative commercial PR antibodies and found that each lacked the specificity and/or sensitivity to immunohistochemically label PR-expressing cells in paraformaldehyde-fixed female mouse brain sections. As a result, we developed and validated a new custom RC269 PR antibody, directed against the same 533-547 amino acid sequence of the human PR as the discontinued A0098 DAKO PR antibody. Immunohistochemical application of the RC269 PR antibody on paraformaldehyde-fixed mouse brain sections resulted in nuclear PR labeling that was highly distinguishable from background, specific to its antigen, highly regulated by estradiol, matched the known distribution of PR protein expression in the female mouse hypothalamus, and nearly identical to that of the discontinued A0098 DAKO PR antibody. In summary, the RC269 PR antibody is a specific and sensitive antibody to immunohistochemically visualize PR-expressing cells in the mouse brain.
Collapse
Affiliation(s)
- Amy Ruddenklau
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Kelly Glendining
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Melanie Prescott
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Rebecca E Campbell
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| |
Collapse
|
14
|
Ma H, Hong WS, Chen SX. A progestin regulates the prostaglandin pathway in the neuroendocrine system in female mudskipper Boleophthalmus pectinirostris. J Steroid Biochem Mol Biol 2023; 231:106300. [PMID: 36990161 DOI: 10.1016/j.jsbmb.2023.106300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 02/02/2023] [Accepted: 02/23/2023] [Indexed: 03/30/2023]
Abstract
Sex hormones regulate the reproductive cycle through brain-pituitary axis, but the molecular mechanism is still enigmatic. In the reproductive season, the mudskipper Boleophthalmus pectinirostris possesses a semilunar periodicity spawning rhythm, which coincides with the semilunar periodicity variations in 17α-hydroxyprogesterone, the precursor of 17α,20β-dihydroxy-4-pregnen-3-one (DHP), a sexual progestin in teleosts. In the present study, we investigated in vitro the brain transcriptional differences between DHP-treated tissues and control groups using RNA-seq. Differential expression analysis revealed that 2700 genes significantly differentially expressed, including 1532 up-regulated and 1168 down-regulated genes. The majority of prostaglandin pathway-related genes were dramatically up-regulated, especially the prostaglandin receptor 6 (ptger6). Tissue distribution analysis revealed that ptger6 gene was ubiquitously expressed. In situ hybridization results showed that ptger6, nuclear progestin receptor (pgr), and DHP-induced c-fos mRNA were co-expressed in the ventral telencephalic area, the ventral nucleus of ventral telencephalic area, the anterior part of parvocellular preoptic nucleus, the magnocellular part of magnocellular preoptic nucleus, the ventral zone of periventricular hypothalamus, the anterior tubercular nucleus, the periventricular nucleus of posterior tuberculum, and the torus longitudinalis. DHP significantly enhanced promoter activities of ptger6 via Pgr. Together, this study suggested that DHP regulates the prostaglandin pathway in the neuroendocrine system of teleost fish.
Collapse
Affiliation(s)
- He Ma
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, China
| | - Wang Shu Hong
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, China; State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, Xiamen University, Xiamen, 361102, China
| | - Shi Xi Chen
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, 361102, China; State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, Xiamen University, Xiamen, 361102, China.
| |
Collapse
|
15
|
Moore AM. Impaired steroid hormone feedback in polycystic ovary syndrome: Evidence from preclinical models for abnormalities within central circuits controlling fertility. Clin Endocrinol (Oxf) 2022; 97:199-207. [PMID: 35349177 PMCID: PMC11289760 DOI: 10.1111/cen.14711] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/09/2022] [Accepted: 02/18/2022] [Indexed: 12/24/2022]
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrinopathy and cause of infertility in women of reproductive age worldwide. Despite diagnostic features of anovulation, polycystic ovarian morphology, and high androgen secretion indicating the syndrome are the result of ovarian dysfunction, alterations to central neuroendocrine circuits that control reproductive capacity may drive PCOS symptoms. Resistance of gonadotrophin-releasing hormone (GnRH) neurons in the hypothalamus to inhibition by sex steroid hormone-negative feedback leads to a rapid frequency of pulsatile gonadotrophin secretion, which, in turn, drives the ovarian features of the disease. As GnRH neurons do not express steroid hormone receptors, impaired negative feedback is hypothesized to occur within an upstream network that controls GnRH pulse generation. This review will discuss the latest work from preclinical animal models of PCOS used to dissect the specific central mechanisms involved in impaired steroid hormone feedback. In particular, this review will focus on research that indicates neurons in the arcuate nucleus of the hypothalamus that express Kisspeptin, Neurokinin B and Dynorphin (KNDy cells) or γ-aminobutyric acid are targets of androgen-mediated impairment of steroid hormone feedback. Finally, this review will explore the development of therapeutic agents targeting neurons that control LH pulse frequency to resolve PCOS symptoms in the clinic.
Collapse
Affiliation(s)
- Aleisha M Moore
- Department of Biological Sciences, Brain Health Research Institute, Kent State University, Kent, Ohio, USA
| |
Collapse
|
16
|
Paiva L, Silva M, Carrasco R, Ratto MH. The ovulatory and luteotropic actions of the male-derived beta-nerve growth factor in South American camelids. Anim Front 2022; 12:87-94. [PMID: 35974784 PMCID: PMC9374510 DOI: 10.1093/af/vfac037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Luis Paiva
- Escuela de Medicina Veterinaria, Facultad de Agronomía e Ingeniería Forestal, Facultad de Ciencias Biológicas y Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mauricio Silva
- Departamento de Medicina Veterinaria y Salud Pública, Núcleo de Investigación en Producción Agroalimentaria, Facultad de Recursos Naturales, Universidad Católica de Temuco, Temuco, Chile
| | - Rodrigo Carrasco
- Department of Chemistry, College of Arts and Science, University of Saskatchewan, Saskatoon, SK,Canada
| | - Marcelo Héctor Ratto
- Instituto de Ciencia Animal, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| |
Collapse
|
17
|
Moenter SM, Evans NP. Gonadotropin-releasing hormone (GnRH) measurements in pituitary portal blood: A history. J Neuroendocrinol 2022; 34:e13065. [PMID: 34918405 PMCID: PMC9200367 DOI: 10.1111/jne.13065] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/29/2021] [Accepted: 11/16/2021] [Indexed: 01/18/2023]
Abstract
Much about the neuroendocrine control of reproduction is inferred from changes in the episodic release of luteinizing hormone (LH), as measured in samples of peripheral blood. This, however, assumes that LH precisely mirrors gonadotropin-releasing hormone (GnRH) release from the hypothalamus. Because GnRH is not measurable in peripheral blood, characterization of the relationship between these two hormones required the simultaneous measurement of GnRH and LH in pituitary portal and peripheral blood, respectively. Here, we review the history of why and how portal blood collection was developed, the aspects of the true output of the central component of the hypothalamic-pituitary-gonadal axis that this methodology helped clarify, and conditions under which the pituitary fails to serve as an adequate bioassay for the release pattern of GnRH.
Collapse
Affiliation(s)
- Suzanne M. Moenter
- Departments of Molecular & Integrative Physiology, Internal Medicine, Obstetrics & Gynecology, and the Reproductive Sciences Program, University of Michigan, Ann Arbor, Michigan, USA
| | - Neil P. Evans
- Institute of Biodiversity Animal Health and Comparative Medicine, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
18
|
Kim SH, Lundgren JA, Patrie JT, Burt Solorzano CM, McCartney CR. Acute progesterone feedback on gonadotropin secretion is not demonstrably altered in estradiol-pretreated women with polycystic ovary syndrome. Physiol Rep 2022; 10:e15233. [PMID: 35384387 PMCID: PMC8981178 DOI: 10.14814/phy2.15233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 02/23/2022] [Accepted: 02/28/2022] [Indexed: 06/14/2023] Open
Abstract
Women with polycystic ovary syndrome (PCOS) demonstrate gonadotropin-releasing hormone (GnRH) pulse generator resistance to suppression with 7 days of progesterone and estradiol administration. It remains unknown whether such women demonstrate impairments in acute progesterone negative feedback on LH pulse frequency or progesterone positive feedback on gonadotropin release. This was a randomized, double-blind, placebo-controlled crossover study designed to test the hypothesis that acute, progesterone-related suppression of LH pulse frequency and progesterone-related augmentation of gonadotropin release are impaired in PCOS. Twelve normally cycling women and 12 women with PCOS completed study. Volunteers were pretreated with transdermal estradiol (0.2 mg/day) for 3 days and then underwent a frequent blood sampling study (20:00-20:00 h), during which they received micronized progesterone (100 mg) or placebo at 06:00 h. In a second study admission, volunteers received the intervention they did not receive during the first admission, but the protocol was otherwise identical. The primary outcome measures were LH secretory characteristics and circulating gonadotropin concentrations. Exogenous progesterone did not reduce LH pulse frequency in either group. Mean LH, pulsatile LH secretion, LH pulse mass, and mean FSH increased more with progesterone compared to placebo in both groups. Although trends toward less pronounced changes in LH pulse mass and pulsatile LH secretion were observed in the PCOS group, these differences were not statistically significant. In summary, exogenous progesterone did not suppress LH pulse frequency within 12 hours in estradiol-pretreated women, and the positive feedback effect of progesterone on gonadotropin release was not demonstrably impaired in PCOS. NEW & NOTEWORTHY: This study indicated that exogenous progesterone does not reduce LH pulse frequency within 12 h in women with PCOS, but progesterone acutely increased gonadotropin in these women. This study suggested that progesterone-related augmentation of gonadotropin release may be impaired in PCOS compared to normally cycling women, but this finding was not statistically significant.
Collapse
Affiliation(s)
- Su Hee Kim
- Center for Research in ReproductionUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
- Division of EndocrinologyDepartment of MedicineUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
| | - Jessica A. Lundgren
- Center for Research in ReproductionUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
- Division of EndocrinologyDepartment of MedicineUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
| | - James T. Patrie
- Department of Public Health SciencesUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
| | - Christine M. Burt Solorzano
- Center for Research in ReproductionUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
- Division of EndocrinologyDepartment of PediatricsUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
| | - Christopher R. McCartney
- Center for Research in ReproductionUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
- Division of EndocrinologyDepartment of MedicineUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
| |
Collapse
|
19
|
Silva MSB, Campbell RE. Polycystic Ovary Syndrome and the Neuroendocrine Consequences of Androgen Excess. Compr Physiol 2022; 12:3347-3369. [PMID: 35578968 DOI: 10.1002/cphy.c210025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a major endocrine disorder strongly associated with androgen excess and frequently leading to female infertility. Although classically considered an ovarian disease, altered neuroendocrine control of gonadotropin-releasing hormone (GnRH) neurons in the brain and abnormal gonadotropin secretion may underpin PCOS presentation. Defective regulation of GnRH pulse generation in PCOS promotes high luteinizing hormone (LH) pulsatile secretion, which in turn overstimulates ovarian androgen production. Early and emerging evidence from preclinical models suggests that maternal androgen excess programs abnormalities in developing neuroendocrine circuits that are associated with PCOS pathology, and that these abnormalities are sustained by postpubertal elevation of endogenous androgen levels. This article will discuss experimental evidence, from the clinic and in preclinical animal models, that has significantly contributed to our understanding of how androgen excess influences the assembly and maintenance of neuroendocrine impairments in the female brain. Abnormal central gamma-aminobutyric acid (GABA) signaling has been identified in both patients and preclinical models as a possible link between androgen excess and elevated GnRH/LH secretion. Enhanced GABAergic innervation and drive to GnRH neurons is suspected to contribute to the pathogenesis and early manifestation of neuroendocrine derangement in PCOS. Accordingly, this article also provides an overview of GABA regulation of GnRH neuron function from prenatal development to adulthood to discuss possible avenues for future discovery research and therapeutic interventions. © 2022 American Physiological Society. Compr Physiol 12:3347-3369, 2022.
Collapse
Affiliation(s)
- Mauro S B Silva
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Rebecca E Campbell
- Centre for Neuroendocrinology, Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
20
|
Diviccaro S, Cioffi L, Falvo E, Giatti S, Melcangi RC. Allopregnanolone: An overview on its synthesis and effects. J Neuroendocrinol 2022; 34:e12996. [PMID: 34189791 PMCID: PMC9285581 DOI: 10.1111/jne.12996] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 05/20/2021] [Accepted: 05/26/2021] [Indexed: 12/23/2022]
Abstract
Allopregnanolone, a 3α,5α-progesterone metabolite, acts as a potent allosteric modulator of the γ-aminobutyric acid type A receptor. In the present review, the synthesis of this neuroactive steroid occurring in the nervous system is discussed with respect to physiological and pathological conditions. In addition, its physiological and neuroprotective effects are also reported. Interestingly, the levels of this neuroactive steroid, as well as its effects, are sex-dimorphic, suggesting a possible gender medicine based on this neuroactive steroid for neurological disorders. However, allopregnanolone presents low bioavailability and extensive hepatic metabolism, limiting its use as a drug. Therefore, synthetic analogues or a different therapeutic strategy able to increase allopregnanolone levels have been proposed to overcome any pharmacokinetic issues.
Collapse
Affiliation(s)
- Silvia Diviccaro
- Dipartimento di Scienze Farmacologiche e BiomolecolariUniversità degli Studi di MilanoMilanoItaly
| | - Lucia Cioffi
- Dipartimento di Scienze Farmacologiche e BiomolecolariUniversità degli Studi di MilanoMilanoItaly
| | - Eva Falvo
- Dipartimento di Scienze Farmacologiche e BiomolecolariUniversità degli Studi di MilanoMilanoItaly
| | - Silvia Giatti
- Dipartimento di Scienze Farmacologiche e BiomolecolariUniversità degli Studi di MilanoMilanoItaly
| | - Roberto Cosimo Melcangi
- Dipartimento di Scienze Farmacologiche e BiomolecolariUniversità degli Studi di MilanoMilanoItaly
| |
Collapse
|
21
|
Watanabe Y, Prescott M, Campbell RE, Jasoni CL. Prenatal androgenization causes expression changes of progesterone and androgen receptor mRNAs in the arcuate nucleus of female mice across development. J Neuroendocrinol 2021; 33:e13058. [PMID: 34748236 DOI: 10.1111/jne.13058] [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: 02/26/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 11/27/2022]
Abstract
Prenatal exposure to excess androgens is associated with the development of polycystic ovary syndrome (PCOS). In prenatally androgenised (PNA) mice, a model of PCOS, progesterone receptor (PR) protein expression is reduced in arcuate nucleus (ARC) GABA neurons. This suggests a mechanism for PCOS-related impaired steroid hormone feedback and implicates androgen excess with respect to inducing transcriptional repression of the PR-encoding gene Pgr in the ARC. However, the androgen sensitivity of ARC neurons and the relative gene expression of PRs over development and following prenatal androgen exposure remain unknown. Here, we used a quantitative reverse transcriptase-polymerase chain reaction (RT-qPCR) of microdissected ARC to determine the relative androgen receptor (Ar) and progesterone receptor (Pgr) gene expression in PNA and control mice at five developmental timepoints. In a two-way analysis of variance, none of the genes examined showed expression changes with a statistically significant interaction between treatment and age, although PgrA showed a borderline interaction. For all genes, there was a statistically significant main effect of age on expression levels, reflecting a general increase in expression with increasing age, regardless of treatment. For PgrB and Ar, there was a statistically significant main effect of treatment, indicating a change in expression following PNA (increased for PgrB and decreased for Ar), regardless of age. For PgrA, there was a borderline main effect of treatment, suggesting a possible change in expression following PNA, regardless of age. PgrAB gene expression changes showed no significant main effect of treatment. We additionally examined androgen and progesterone responsiveness specifically in P60 ARC GABA neurons using RNAScope® (Advanced Cell Diagnostics, Inc.) in situ hybridization. This analysis revealed that Pgr and Ar were expressed in the majority of ARC GABA neurons in normal adult females. However, our RNAScope® analysis did not show significant changes in Pgr or Ar expression within ARC GABA neurons following PNA. Lastly, because GABA drive to gonadotropin-releasing hormone neurons is increased in PNA, we hypothesised that PNA mice would show increased expression of glutamic acid decarboxylase (GAD), the rate-limiting enzyme in GABA production. However, the RT-qPCR showed that the expression of GAD encoding genes (Gad1 and Gad2) was unchanged in adult PNA mice compared to controls. Our findings indicate that PNA treatment can impact Pgr and Ar mRNA expression in adulthood. This may reflect altered circulating steroid hormones in PNA mice or PNA-induced epigenetic changes in the regulation of Pgr and Ar gene expression in ARC neurons.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Arcuate Nucleus of Hypothalamus/growth & development
- Arcuate Nucleus of Hypothalamus/metabolism
- Embryo, Mammalian
- Female
- Gene Expression Regulation, Developmental
- Growth and Development/genetics
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Pregnancy
- Prenatal Exposure Delayed Effects/genetics
- Prenatal Exposure Delayed Effects/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Receptors, Progesterone/genetics
- Receptors, Progesterone/metabolism
- Virilism/embryology
- Virilism/genetics
- Virilism/metabolism
Collapse
Affiliation(s)
- Yugo Watanabe
- Department of Anatomy, Centre for Neuroendocrinology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Melanie Prescott
- Department of Physiology, Centre for Neuroendocrinology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Rebecca E Campbell
- Department of Physiology, Centre for Neuroendocrinology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Christine L Jasoni
- Department of Anatomy, Centre for Neuroendocrinology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| |
Collapse
|
22
|
Voliotis M, Li XF, De Burgh RA, Lass G, Ivanova D, McIntyre C, O'Byrne K, Tsaneva-Atanasova K. Modulation of pulsatile GnRH dynamics across the ovarian cycle via changes in the network excitability and basal activity of the arcuate kisspeptin network. eLife 2021; 10:e71252. [PMID: 34787076 PMCID: PMC8651288 DOI: 10.7554/elife.71252] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 11/16/2021] [Indexed: 11/13/2022] Open
Abstract
Pulsatile GnRH release is essential for normal reproductive function. Kisspeptin secreting neurons found in the arcuate nucleus, known as KNDy neurons for co-expressing neurokinin B, and dynorphin, drive pulsatile GnRH release. Furthermore, gonadal steroids regulate GnRH pulsatile dynamics across the ovarian cycle by altering KNDy neurons' signalling properties. However, the precise mechanism of regulation remains mostly unknown. To better understand these mechanisms, we start by perturbing the KNDy system at different stages of the estrous cycle using optogenetics. We find that optogenetic stimulation of KNDy neurons stimulates pulsatile GnRH/LH secretion in estrous mice but inhibits it in diestrous mice. These in vivo results in combination with mathematical modelling suggest that the transition between estrus and diestrus is underpinned by well-orchestrated changes in neuropeptide signalling and in the excitability of the KNDy population controlled via glutamate signalling. Guided by model predictions, we show that blocking glutamate signalling in diestrous animals inhibits LH pulses, and that optic stimulation of the KNDy population mitigates this inhibition. In estrous mice, disruption of glutamate signalling inhibits pulses generated via sustained low-frequency optic stimulation of the KNDy population, supporting the idea that the level of network excitability is critical for pulse generation. Our results reconcile previous puzzling findings regarding the estradiol-dependent effect that several neuromodulators have on the GnRH pulse generator dynamics. Therefore, we anticipate our model to be a cornerstone for a more quantitative understanding of the pathways via which gonadal steroids regulate GnRH pulse generator dynamics. Finally, our results could inform useful repurposing of drugs targeting the glutamate system in reproductive therapy.
Collapse
Affiliation(s)
- Margaritis Voliotis
- Department of Mathematics and Living Systems Institute, College of Engineering, Mathematics and Physical Sciences, University of ExeterExeterUnited Kingdom
| | - Xiao Feng Li
- Department of Women and Children’s Health, School of Life Course Sciences, King’s College LondonLondonUnited Kingdom
| | - Ross Alexander De Burgh
- Department of Women and Children’s Health, School of Life Course Sciences, King’s College LondonLondonUnited Kingdom
| | - Geffen Lass
- Department of Women and Children’s Health, School of Life Course Sciences, King’s College LondonLondonUnited Kingdom
| | - Deyana Ivanova
- Department of Women and Children’s Health, School of Life Course Sciences, King’s College LondonLondonUnited Kingdom
| | - Caitlin McIntyre
- Department of Women and Children’s Health, School of Life Course Sciences, King’s College LondonLondonUnited Kingdom
| | - Kevin O'Byrne
- Department of Women and Children’s Health, School of Life Course Sciences, King’s College LondonLondonUnited Kingdom
| | - Krasimira Tsaneva-Atanasova
- Department of Mathematics and Living Systems Institute, College of Engineering, Mathematics and Physical Sciences, University of ExeterExeterUnited Kingdom
| |
Collapse
|
23
|
Lindo AN, Thorson JF, Bedenbaugh MN, McCosh RB, Lopez JA, Young SA, Meadows LJ, Bowdridge EC, Fergani C, Freking BA, Lehman MN, Hileman SM, Lents CA. Localization of kisspeptin, NKB, and NK3R in the hypothalamus of gilts treated with the progestin altrenogest. Biol Reprod 2021; 105:1056-1067. [PMID: 34037695 DOI: 10.1093/biolre/ioab103] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 05/17/2021] [Accepted: 05/21/2021] [Indexed: 11/12/2022] Open
Abstract
Mechanisms in the brain controlling secretion of gonadotropin hormones in pigs, particularly luteinizing hormone (LH), are poorly understood. Kisspeptin is a potent LH stimulant that is essential for fertility in many species, including pigs. Neurokinin B (NKB) acting through neurokinin 3 receptor (NK3R) is involved in kisspeptin-stimulated LH release, but organization of NKB and NK3R within the porcine hypothalamus is unknown. Hypothalamic tissue from ovariectomized (OVX) gilts was used to determine the distribution of immunoreactive kisspeptin, NKB, and NK3R cells in the arcuate nucleus (ARC). Almost all kisspeptin neurons coexpressed NKB in the porcine ARC. Immunostaining for NK3R was distributed throughout the preoptic area (POA) and in several hypothalamic areas including the periventricular and retrochiasmatic areas but was not detected within the ARC. There was no colocalization of NK3R with gonadotropin-releasing hormone (GnRH), but NK3R-positive fibers in the POA were in close apposition to GnRH neurons. Treating OVX gilts with the progestin altrenogest decreased LH pulse frequency and reduced mean circulating concentrations of LH compared with OVX control gilts (P < 0.01), but the number of kisspeptin and NKB cells in the ARC did not differ between treatments. The neuroanatomical arrangement of kisspeptin, NKB, and NK3R within the porcine hypothalamus confirm they are positioned to stimulate GnRH and LH secretion in gilts, though differences with other species exist. Altrenogest suppression of LH secretion in the OVX gilt does not appear to involve decreased peptide expression of kisspeptin or NKB.
Collapse
Affiliation(s)
- Ashley N Lindo
- Department of Physiology and Pharmacology and Department of Neuroscience, West Virginia University, Morgantown, WV, USA
| | | | - Michelle N Bedenbaugh
- Department of Physiology and Pharmacology and Department of Neuroscience, West Virginia University, Morgantown, WV, USA
| | - Richard B McCosh
- Department of Physiology and Pharmacology and Department of Neuroscience, West Virginia University, Morgantown, WV, USA
| | - Justin A Lopez
- Department of Physiology and Pharmacology and Department of Neuroscience, West Virginia University, Morgantown, WV, USA
| | - Samantha A Young
- Department of Physiology and Pharmacology and Department of Neuroscience, West Virginia University, Morgantown, WV, USA
| | - Lanny J Meadows
- Department of Physiology and Pharmacology and Department of Neuroscience, West Virginia University, Morgantown, WV, USA
| | - Elizabeth C Bowdridge
- Department of Physiology and Pharmacology and Department of Neuroscience, West Virginia University, Morgantown, WV, USA
| | - Chrysanthi Fergani
- Department of Neurobiology and Anatomical Sciences, The University of Mississippi Medical Center, Jackson, Miss., USA
| | | | - Michael N Lehman
- Department of Biological Sciences and the Brain Health Research Institute, Kent State University, Kent, OH, USA
| | - Stanley M Hileman
- Department of Physiology and Pharmacology and Department of Neuroscience, West Virginia University, Morgantown, WV, USA
| | - Clay A Lents
- USDA, ARS, U.S. Meat Animal Research Center, Clay Center, NE, USA
| |
Collapse
|
24
|
Dinh T, Gangestad SW, Thompson ME, Tomiyama AJ, Fessler DMT, Robertson TE, Haselton MG. Endocrinological effects of social exclusion and inclusion: Experimental evidence for adaptive regulation of female fecundity. Horm Behav 2021; 130:104934. [PMID: 33476675 DOI: 10.1016/j.yhbeh.2021.104934] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 12/10/2020] [Accepted: 01/07/2021] [Indexed: 11/30/2022]
Abstract
When current conditions are probabilistically less suitable for successful reproduction than future conditions, females may prevent or delay reproduction until conditions improve. Throughout human evolution, social support was likely crucial to female reproductive success. Women may thus have evolved fertility regulation systems sensitive to cues from the social environment. However, current understanding of how psychological phenomena might affect female ovarian function is limited. In this study, we examined whether cues of reduced social support-social ostracism-impact women's hormone production. Following an in-lab group bonding task, women were randomly assigned to a social exclusion (n = 88) or social inclusion (n = 81) condition. After social exclusion, women with low background levels of social support experienced a decrease in estradiol relative to progesterone. In contrast, socially-included women with low background social support experienced an increase in estradiol relative to progesterone. Hormonal changes in both conditions occurred specifically when women were in their mid-to-late follicular phase, when baseline estradiol is high and progesterone is low. Follow-up analyses revealed that these changes were primarily driven by changes in progesterone, consistent with existing evidence for disruption of ovarian function following adrenal release of follicular-phase progesterone. Results offer support for a potential mechanism by which fecundity could respond adaptively to the loss or lack of social support.
Collapse
Affiliation(s)
- Tran Dinh
- Department of Psychology, University of New Mexico, Albuquerque, NM, USA; Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Steven W Gangestad
- Department of Psychology, University of New Mexico, Albuquerque, NM, USA
| | | | - A Janet Tomiyama
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA; Bedari Kindness Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Daniel M T Fessler
- Department of Anthropology, University of California, Los Angeles, Los Angeles, CA, USA; Bedari Kindness Institute, University of California, Los Angeles, Los Angeles, CA, USA; Center for Behavior, Evolution, & Culture, University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Martie G Haselton
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA; Department of Communication, University of California, Los Angeles, CA, USA
| |
Collapse
|
25
|
Papadaki M, Kaitetzidou E, Mylonas CC, Sarropoulou E. Non-coding RNA Expression Patterns of Two Different Teleost Gonad Maturation Stages. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2020; 22:683-695. [PMID: 32876760 DOI: 10.1007/s10126-020-09991-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/19/2020] [Indexed: 06/11/2023]
Abstract
Non-coding RNAs (ncRNAs) are involved in several different regulatory pathways including reproduction. In teleost fish, efficacious reproduction is heavily dependent on the completion of the reproductive cycle. The presence of ncRNA, however, and their expression dynamics and putative regulatory role in mature and immature gonads have not yet been extensively explored. Therefore, the abundance of ncRNAs in mature and immature female sharpsnout seabream (Diplodus puntazzo) was investigated. The sharpsnout seabream is a rudimentary hermaphrodite which, in captivity, displays dysfunctions in the gonad maturation process. Our analyses revealed a gonad specific read length distribution with two main peaks representing miRNAs (21-26 nt) and PIWI RNA (27-34 nt). Besides, distinct expression patterns for several ncRNA biotypes including microRNAs (miRNAs), PIWI RNAs (piRNAs), and ribosomal RNAs (rRNAs) were detected. Identified miRNA accounted to 938, corresponding to ~ 13% of obtained transcripts. Among the differential expressed ncRNAs, 10 (~ 7%) were annotated as miRNA, out of which 2 were found in higher abundance in immature gonads (miR-125c and miR-24) and 8 (miR-451, miR-7a, miR-122-1, miR190a, miR129, ENSGACT00000029608, ENSGACT00000029489, and ENSGACT00000029667) were found to be higher expressed in mature gonads. Putative miRNA targets, including long non-coding RNAs (lncRNAs) and genes, are proposed. Target genes are involved in several processes of fish oocyte development, such as steroidogenesis, proteolysis, and apoptosis, and may explain hormone regulation. This study demonstrates a gonad maturation biased ncRNA profile which in turn may support the role of ncRNAs in ovarian physiology and reproductive performance of fish, stressing the specific function of each RNA biotype in oocyte development.
Collapse
Affiliation(s)
- Maria Papadaki
- Institute of Marine Biology, Biotechnology and Aquaculture, Hellenic Centre for Marine Research, P.O. Box 2214, 71003, Iraklion, Crete, Greece
- Department of Biology, University of Crete, P.O. Box 2208, 71409, Iraklion, Crete, Greece
| | - Elisavet Kaitetzidou
- Institute of Marine Biology, Biotechnology and Aquaculture, Hellenic Centre for Marine Research, P.O. Box 2214, 71003, Iraklion, Crete, Greece
| | - Constantinos C Mylonas
- Institute of Marine Biology, Biotechnology and Aquaculture, Hellenic Centre for Marine Research, P.O. Box 2214, 71003, Iraklion, Crete, Greece
| | - Elena Sarropoulou
- Institute of Marine Biology, Biotechnology and Aquaculture, Hellenic Centre for Marine Research, P.O. Box 2214, 71003, Iraklion, Crete, Greece.
| |
Collapse
|
26
|
Single-Cell Gene Profiling Reveals Social Status-Dependent Modulation of Nuclear Hormone Receptors in GnRH Neurons in a Male Cichlid Fish. Int J Mol Sci 2020; 21:ijms21082724. [PMID: 32326396 PMCID: PMC7215790 DOI: 10.3390/ijms21082724] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 01/16/2020] [Accepted: 01/17/2020] [Indexed: 12/17/2022] Open
Abstract
Gonadotropin-releasing hormone (GnRH) is essential for the initiation and maintenance of reproductive functions in vertebrates. To date, three distinct paralogue lineages, GnRH1, GnRH2, and GnRH3, have been identified with different functions and regulatory mechanisms. Among them, hypothalamic GnRH1 neurons are classically known as the hypophysiotropic form that is regulated by estrogen feedback. However, the mechanism of action underlying the estrogen-dependent regulation of GnRH1 has been debated, mainly due to the coexpression of low levels of estrogen receptor (ER) genes. In addition, the role of sex steroids in the modulation of GnRH2 and GnRH3 neurons has not been fully elucidated. Using single-cell real-time PCR, we revealed the expression of genes for estrogen, androgen, glucocorticoid, thyroid, and xenobiotic receptors in GnRH1, GnRH2, and GnRH3 neurons in the male Nile tilapia Oreochromis niloticus. We further quantified expression levels of estrogen receptor genes (ERα, ERβ, and ERγ) in three GnRH neuron types in male tilapia of two different social statuses (dominant and subordinate) at the single cell level. In dominant males, GnRH1 mRNA levels were positively proportional to ERγ mRNA levels, while in subordinate males, GnRH2 mRNA levels were positively proportional to ERβ mRNA levels. These results indicate that variations in the expression of nuclear receptors (and possibly steroid sensitivities) among individual GnRH cells may facilitate different physiological processes, such as the promotion of reproductive activities through GnRH1 neurons, and the inhibition of feeding and sexual behaviors through GnRH2 neurons.
Collapse
|
27
|
Herbison AE. A simple model of estrous cycle negative and positive feedback regulation of GnRH secretion. Front Neuroendocrinol 2020; 57:100837. [PMID: 32240664 DOI: 10.1016/j.yfrne.2020.100837] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 03/19/2020] [Accepted: 03/19/2020] [Indexed: 12/19/2022]
Abstract
The gonadal steroids estradiol and progesterone exert critical suppressive and stimulatory actions upon the brain to control gonadotropin-releasing hormone (GnRH) release that drives the estrous/menstrual cycle. A simple model for understanding these interactions is proposed in which the activity of the "GnRH pulse generator" is restrained by post-ovulation progesterone secretion to bring about the estrus/luteal phase slowing of pulsatile gonadotropin release, while the activity of the "GnRH surge generator" is primed by the rising follicular phase levels of estradiol to generate the pre-ovulatory surge. The physiological fluctuations in estradiol levels across the cycle are considered to clamp the GnRH pulse generator output at a constant level. Independent pulse and surge generator circuitries regulate the excitability of different compartments of the GnRH neuron. As such, GnRH secretion through the cycle is determined simply by the summed influence of the estradiol-clamped, progesterone-regulated pulse and estradiol-regulated surge generators on the GnRH neuron.
Collapse
Affiliation(s)
- Allan E Herbison
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin 9054, New Zealand.
| |
Collapse
|
28
|
Giatti S, Diviccaro S, Serafini MM, Caruso D, Garcia-Segura LM, Viviani B, Melcangi RC. Sex differences in steroid levels and steroidogenesis in the nervous system: Physiopathological role. Front Neuroendocrinol 2020; 56:100804. [PMID: 31689419 DOI: 10.1016/j.yfrne.2019.100804] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/10/2019] [Accepted: 10/30/2019] [Indexed: 12/13/2022]
Abstract
The nervous system, in addition to be a target for steroid hormones, is the source of a variety of neuroactive steroids, which are synthesized and metabolized by neurons and glial cells. Recent evidence indicates that the expression of neurosteroidogenic proteins and enzymes and the levels of neuroactive steroids are different in the nervous system of males and females. We here summarized the state of the art of neuroactive steroids, particularly taking in consideration sex differences occurring in the synthesis and levels of these molecules. In addition, we discuss the consequences of sex differences in neurosteroidogenesis for the function of the nervous system under healthy and pathological conditions and the implications of neuroactive steroids and neurosteroidogenesis for the development of sex-specific therapeutic interventions.
Collapse
Affiliation(s)
- Silvia Giatti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Silvia Diviccaro
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Melania Maria Serafini
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Donatella Caruso
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Luis Miguel Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Barbara Viviani
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Roberto C Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy.
| |
Collapse
|
29
|
Leonardi CEP, Dias FCF, Adams GP, Araujo ER, Singh J. Kisspeptin induces ovulation in heifers under low plasma progesterone concentrations. Theriogenology 2020; 141:26-34. [PMID: 31494459 DOI: 10.1016/j.theriogenology.2019.08.033] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 08/14/2019] [Accepted: 08/28/2019] [Indexed: 11/23/2022]
Abstract
The objective of the present study was to compare the effect of a single versus multiple doses of a 10-amino acid fragment of human (hKp) or murine (mKp) kisspeptin on LH secretion and the fate of the dominant follicle. In all experiments, a new wave was induced (Day 0) by ultrasound-guided ablation of >5 mm follicles, a progesterone device (CIDR) was placed in the vagina, animals given prostaglandin F2α analog im on Day 3.5 and 4, and hKp or mKp treatment given on Day 6. The experimental design maintained growth and ovulatory potential of the dominant follicle for 12 days and allowed hypothesis testing during the low-progesterone period (plasma progesterone ≤1.8 ng/ml on Day 6) wherein spontaneous wave emergence and ovulation did not occur between Day 6 and Day 12. In Experiment 1, heifers (n = 10/group) were given single iv dose of 45 mg hKp, 45 mg mKp, or 2 ml normal saline (control). Post-treatment plasma LH concentrations from 15 to 90 min were higher (P < 0.01) in hKp group than in the mKp and control groups. Two heifers ovulated in hKp group versus none in other groups. In Experiment 2, heifers (n = 6/group) were given 45 mg hKp over a 2 h period divided into multiple iv doses treatments or 2 ml normal saline (control). Post-treatment plasma LH concentrations were higher (P < 0.01) in all hKp treatment groups than in the control group. The ovulation rate was higher (P = 0.06) after hKp treatments (11/18) than in the control group (0/6). In Experiment 3, heifers (n = 6/group) were given 45 mg mKp over a 2 h period divided into multiple iv doses treatments or a single iv dose of gonadorelin acetate (positive control). Plasma LH concentration was higher (P < 0.01) and the ovulation rate was greater (P = 0.01) in the GnRH group (5/6) than mKp groups (1/12). In summary, hKp was more effective to induce ovulation than mKp. Human kisspeptin-10 given over a 2 h period induced ovulations at a rate similar to that of GnRH treatment in heifers under a low plasma progesterone state.
Collapse
Affiliation(s)
- Carlos E P Leonardi
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Fernanda C F Dias
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Gregg P Adams
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Estela R Araujo
- Department of Pathobiological Sciences, University of Wisconsin, Madison, USA
| | - Jaswant Singh
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.
| |
Collapse
|
30
|
Progestogen profiling in plasma during the estrous cycle in cattle using an LC-MS based approach. Theriogenology 2019; 142:376-383. [PMID: 31708192 DOI: 10.1016/j.theriogenology.2019.10.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 10/02/2019] [Accepted: 10/08/2019] [Indexed: 11/23/2022]
Abstract
In many mammalian species, corpus luteum derived progesterone (P4) is the main functional gestagen during the estrous cycle and pregnancy. P4 can be metabolized into various metabolites, of which some are biologically active. While some metabolites target the classical nuclear progesterone receptor (PR), neurosteroids bind the receptors of type A γ-aminobutyric acid (GABAA-r) in the brain. According to the position of reduction within the molecule, metabolites of P4 can be characterized into C20-reduced progestogens (20α-dihydroprogesterone (20α-DHP) and 20β-dihydroprogesterone (20β-DHP)), C3-reduced progestogens (3α-dihydroprogesterone (3α-DHP) and 3β-dihydroprogesterone (3β-DHP)), 5α-reduced progestogens (5α-dihydroprogesterone (5α-DHP), allopregnanolone and isopregnanolone) and 5β-reduced progestogens (5β-dihydroprogesterone (5β-DHP), pregnanolone and epipregnanolone). We questioned whether the reduced progestogens are present in bovine plasma during the estrous cycle and whether their profiles differed from the profile of the common precursor P4 around the time of luteolysis. The analytes were monitored in plasma samples using liquid chromatography mass spectrometry (LC-MS). While progestogens lagged behind the drop of P4 at luteolysis, they followed the profile of P4 during the estrous cycle. The abundance of P4 was predominant followed by allopregnanolone, pregnanolone, epipregnanolone and 20β-DHP. Further studies will need to focus particularly on the period around luteolysis.
Collapse
|
31
|
McQuillan HJ, Han SY, Cheong I, Herbison AE. GnRH Pulse Generator Activity Across the Estrous Cycle of Female Mice. Endocrinology 2019; 160:1480-1491. [PMID: 31083714 DOI: 10.1210/en.2019-00193] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 04/16/2019] [Indexed: 11/19/2022]
Abstract
A subpopulation of kisspeptin neurons located in the arcuate nucleus (ARN) operate as the GnRH pulse generator. The activity of this population of neurons can be monitored in real-time for long periods using kisspeptin neuron-selective GCaMP6 fiber photometry. Using this approach, we find that ARN kisspeptin neurons exhibit brief (∼50 seconds) periods of synchronized activity that precede pulses of LH in intact female mice. The dynamics and frequency of these synchronization episodes (SEs) are stable at approximately one event every 40 minutes throughout metestrus, diestrus, and proestrus, but slow considerably on estrus to occur approximately once every 10 hours. Evaluation of ARN kisspeptin neuron activity across the light-dark transition, including the time of onset of the proestrus LH surge, revealed no changes in SE frequency. Longer 24-hour recordings across proestrus into estrus demonstrated that an abrupt decrease in SEs occurred ∼4 to 5 hours after the onset of the LH surge to reach the low frequency of SEs observed on estrus. The frequency of SEs was stable across the 24-hour period from metestrus to diestrus. Administration of progesterone to diestrus mice resulted in the abrupt slowing of SEs. These observations show that the GnRH pulse generator exhibits an unvarying pattern of activity from metestrus through to the late evening of proestrus, at which time it slows dramatically, likely in response to postovulation progesterone secretion. The GnRH pulse generator maintains a constant frequency of activity across the time of the LH surge, demonstrating that it is not involved directly in surge generation.
Collapse
Affiliation(s)
- H James McQuillan
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Su Young Han
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Isaiah Cheong
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| | - Allan E Herbison
- Centre for Neuroendocrinology and Department of Physiology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand
| |
Collapse
|
32
|
Proietto S, Yankelevich L, Villarreal FM, Inserra PIF, Charif SE, Schmidt AR, Cortasa SA, Corso MC, Di Giorgio NP, Lux-Lantos V, Vitullo AD, Halperin J, Dorfman VB. Pituitary estrogen receptor alpha is involved in luteinizing hormone pulsatility at mid-gestation in the South American plains vizcacha, Lagostomus maximus (Rodentia, Caviomorpha). Gen Comp Endocrinol 2019; 273:40-51. [PMID: 29656043 DOI: 10.1016/j.ygcen.2018.04.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 03/22/2018] [Accepted: 04/02/2018] [Indexed: 11/30/2022]
Abstract
The South American plains vizcacha, Lagostomus maximus, is a caviomorph rodent native from Argentina, Bolivia and Paraguay. It shows peculiar reproductive features like pre-ovulatory follicle recruitment during pregnancy with an ovulatory process at around mid-gestation. We have described the activation of the hypothalamic - pituitary - ovarian (HPO) axis during pregnancy. A progressive decrease of progesterone (P4) at mid-pregnancy elicits the delivery of gonadotropin-releasing hormone (GnRH) with the consequent secretion of follicle stimulating hormone (FSH) and estradiol (E2) followed by luteinizing hormone (LH) release resulting in follicular luteinization and the P4 concentration recover. Pituitary gland is the central regulator of the HPO axis being E2 a key hormone involved in the regulation of its activity. In this work we analyzed the action of E2 on the pituitary response to the GnRH wave as well as its involvement on LH secretion at mid-gestation in L. maximus. The expression of GnRHR at the pituitary pars distalis showed a significant decrease at mid-pregnancy compared to early- and term-gestating females. ERα showed a significant increment from mid-gestation whereas ERβ did not show variations throughout pregnancy; whereas the LH expression in the pituitary pars distalis showed a significant increase at mid-gestation, concordantly with serum LH, which was followed by a decrease at term-gestation with similar values than at early-pregnancy. The number of cells with co-localization of ERα and GnRHR showed a decline at mid-pregnancy related to early- and term-gestation, whereas the cells with co-localization of ERα and LH increased at mid- and term-pregnancy. On the other hand, ex vivo measuring of LH pulsatility showed a significant increment in the total mass of LH delivered at mid-pregnancy followed by a decrease at term-gestation. The stimulation of ERα with the PPT specific agonist induced a significant increment in the total mass of LH released, whereas no changes were determined when ERβ was stimulated with its specific agonist MPP. These results suggest that LH pulsatility rise at mid-pregnancy would be enabled by the increase of E2 acting through ERα.
Collapse
Affiliation(s)
- Sofía Proietto
- 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), Argentina
| | - Lorena Yankelevich
- Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina
| | - Federico Martín Villarreal
- Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina
| | - Pablo Ignacio Felipe 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), Argentina
| | - Santiago Elías 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), Argentina
| | - Alejandro Raúl 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), Argentina
| | - Santiago Andrés 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), Argentina
| | - María Clara Corso
- 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), Argentina
| | - Noelia Paula Di Giorgio
- Laboratorio de Neuroendocrinología, Instituto de Biología y Medicina Experimental (IByME)-CONICET, Ciudad Autónoma de Buenos Aires, Argentina
| | - Victoria Lux-Lantos
- Laboratorio de Neuroendocrinología, Instituto de Biología y Medicina Experimental (IByME)-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), 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), Argentina
| | - Verónica Berta 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), Argentina.
| |
Collapse
|
33
|
Giatti S, Diviccaro S, Panzica G, Melcangi RC. Post-finasteride syndrome and post-SSRI sexual dysfunction: two sides of the same coin? Endocrine 2018; 61:180-193. [PMID: 29675596 DOI: 10.1007/s12020-018-1593-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 04/05/2018] [Indexed: 12/27/2022]
Abstract
Sexual dysfunction is a clinical condition due to different causes including the iatrogenic origin. For instance, it is well known that sexual dysfunction may occur in patients treated with antidepressants like selective serotonin reuptake inhibitors (SSRI). A similar side effect has been also reported during treatment with finasteride, an inhibitor of the enzyme 5alpha-reductase, for androgenetic alopecia. Interestingly, sexual dysfunction persists in both cases after drug discontinuation. These conditions have been named post-SSRI sexual dysfunction (PSSD) and post-finasteride syndrome (PFS). In particular, feeling of a lack of connection between the brain and penis, loss of libido and sex drive, difficulty in achieving an erection and genital paresthesia have been reported by patients of both conditions. It is interesting to note that the incidence of these diseases is probably so far underestimated and their etiopathogenesis is not sufficiently explored. To this aim, the present review will report the state of art of these two different pathologies and discuss, on the basis of the role exerted by three different neuromodulators such as dopamine, serotonin and neuroactive steroids, whether the persistent sexual dysfunction observed could be determined by common mechanisms.
Collapse
Affiliation(s)
- Silvia Giatti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Silvia Diviccaro
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Giancarlo Panzica
- Dipartimento di Neuroscienze "Rita Levi Montalcini", Università degli studi di Torino, Neuroscience Institute Cavallieri Ottolenghi (NICO), Orbassano, Italy
| | - Roberto Cosimo Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
34
|
Nestor CC, Bedenbaugh MN, Hileman SM, Coolen LM, Lehman MN, Goodman RL. Regulation of GnRH pulsatility in ewes. Reproduction 2018; 156:R83-R99. [PMID: 29880718 DOI: 10.1530/rep-18-0127] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 06/07/2018] [Indexed: 01/21/2023]
Abstract
Early work in ewes provided a wealth of information on the physiological regulation of pulsatile gonadotropin-releasing hormone (GnRH) secretion by internal and external inputs. Identification of the neural systems involved, however, was limited by the lack of information on neural mechanisms underlying generation of GnRH pulses. Over the last decade, considerable evidence supported the hypothesis that a group of neurons in the arcuate nucleus that contain kisspeptin, neurokinin B and dynorphin (KNDy neurons) are responsible for synchronizing secretion of GnRH during each pulse in ewes. In this review, we describe our current understanding of the neural systems mediating the actions of ovarian steroids and three external inputs on GnRH pulsatility in light of the hypothesis that KNDy neurons play a key role in GnRH pulse generation. In breeding season adults, estradiol (E2) and progesterone decrease GnRH pulse amplitude and frequency, respectively, by actions on KNDy neurons, with E2 decreasing kisspeptin and progesterone increasing dynorphin release onto GnRH neurons. In pre-pubertal lambs, E2 inhibits GnRH pulse frequency by decreasing kisspeptin and increasing dynorphin release, actions that wane as the lamb matures to allow increased pulsatile GnRH secretion at puberty. Less is known about mediators of undernutrition and stress, although some evidence implicates kisspeptin and dynorphin, respectively, in the inhibition of GnRH pulse frequency by these factors. During the anoestrus, inhibitory photoperiod acting via melatonin activates A15 dopaminergic neurons that innervate KNDy neurons; E2 increases dopamine release from these neurons to inhibit KNDy neurons and suppress the frequency of kisspeptin and GnRH release.
Collapse
Affiliation(s)
- Casey C Nestor
- Department of Animal Science, North Carolina State University, Raleigh, North Carolina, USA
| | - Michelle N Bedenbaugh
- Department of Physiology, Pharmacology and Neuroscience, West Virginia University, Morgantown, West Virginia, USA
| | - Stanley M Hileman
- Department of Physiology, Pharmacology and Neuroscience, West Virginia University, Morgantown, West Virginia, USA
| | - Lique M Coolen
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA.,Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Michael N Lehman
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Robert L Goodman
- Department of Physiology, Pharmacology and Neuroscience, West Virginia University, Morgantown, West Virginia, USA
| |
Collapse
|
35
|
Kim SH, Burt Solorzano CM, McCartney CR. Progesterone administration does not acutely alter LH pulse secretion in the mid-follicular phase in women. Physiol Rep 2018; 6:e13680. [PMID: 29696832 PMCID: PMC5917047 DOI: 10.14814/phy2.13680] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 03/13/2018] [Accepted: 03/16/2018] [Indexed: 11/24/2022] Open
Abstract
It remains unclear how rapidly progesterone suppresses luteinizing hormone (LH) pulse frequency in women. Previous studies suggested that progesterone markedly increases LH pulse amplitude but does not slow LH pulse frequency within 10 h in estradiol‐pretreated women studied during the late follicular phase. However, this experimental paradigm may be a model of preovulatory physiology, and progesterone may have different effects at other times of the cycle. We studied regularly cycling, nonobese women without hyperandrogenism to assess the acute effect of progesterone during the midfollicular phase and in the absence of estradiol pretreatment. The study involved two admissions in separate cycles (cycle days 5–9). For each admission, either oral micronized progesterone (100 mg) or placebo was administered at 0900 h in a randomized, double‐blind fashion. Frequent blood sampling was performed between 0900 and 1900 h to define 10‐h LH pulsatility. Treatment crossover (placebo exchanged for progesterone and vice versa) occurred in a subsequent cycle. After an interim futility analysis, the study was halted after 7 women completed study. Mean progesterone concentrations after placebo and progesterone administration were 0.5 ± 0.1 (mean ± SD) and 6.7 ± 1.6 ng/mL, respectively. Compared to placebo, progesterone was not associated with a significant difference in 10‐h LH pulse frequency (0.79 ± 0.35 vs. 0.77 ± 0.28 pulses/h, P = 1.0) or amplitude (3.6 ± 2.8 vs. 4.3 ± 2.8 IU/L, P = 0.30). This study suggests that LH pulse frequency is not rapidly influenced by progesterone administration during the midfollicular phase.
Collapse
Affiliation(s)
- Su Hee Kim
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia.,Center for Research in Reproduction, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Christine M Burt Solorzano
- Center for Research in Reproduction, University of Virginia School of Medicine, Charlottesville, Virginia.,Division of Endocrinology and Metabolism, Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Christopher R McCartney
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia.,Center for Research in Reproduction, University of Virginia School of Medicine, Charlottesville, Virginia
| |
Collapse
|
36
|
Weems PW, Lehman MN, Coolen LM, Goodman RL. The Roles of Neurokinins and Endogenous Opioid Peptides in Control of Pulsatile LH Secretion. VITAMINS AND HORMONES 2018; 107:89-135. [PMID: 29544644 DOI: 10.1016/bs.vh.2018.01.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
Abstract
Work over the last 15 years on the control of pulsatile LH secretion has focused largely on a set of neurons in the arcuate nucleus (ARC) that contains two stimulatory neuropeptides, critical for fertility in humans (kisspeptin and neurokinin B (NKB)) and the inhibitory endogenous opioid peptide (EOP), dynorphin, and are now known as KNDy (kisspeptin-NKB-dynorphin) neurons. In this review, we consider the role of each of the KNDy peptides in the generation of GnRH pulses and the negative feedback actions of ovarian steroids, with an emphasis on NKB and dynorphin. With regard to negative feedback, there appear to be important species differences. In sheep, progesterone inhibits GnRH pulse frequency by stimulating dynorphin release, and estradiol inhibits pulse amplitude by suppressing kisspeptin. In rodents, the role of KNDy neurons in estrogen negative feedback remains controversial, progesterone may inhibit GnRH via dynorphin, but the physiological significance of this action is unclear. In primates, an EOP, probably dynorphin, mediates progesterone negative feedback, and estrogen inhibits kisspeptin expression. In contrast, there is now compelling evidence from several species that kisspeptin is the output signal from KNDy neurons that drives GnRH release during a pulse and may also act within the KNDy network to affect pulse frequency. NKB is thought to act within this network to initiate each pulse, although there is some redundancy in tachykinin signaling in rodents. In ruminants, dynorphin terminates GnRH secretion at the end of pulse, most likely acting on both KNDy and GnRH neurons, but the data on the role of this EOP in rodents are conflicting.
Collapse
Affiliation(s)
- Peyton W Weems
- Graduate Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS, United States
| | - Michael N Lehman
- University of Mississippi Medical Center, Jackson, MS, United States
| | - Lique M Coolen
- University of Mississippi Medical Center, Jackson, MS, United States
| | | |
Collapse
|
37
|
Hutchens EG, Ramsey KA, Howard LC, Abshire MY, Patrie JT, McCartney CR. Progesterone has rapid positive feedback actions on LH release but fails to reduce LH pulse frequency within 12 h in estradiol-pretreated women. Physiol Rep 2017; 4:4/16/e12891. [PMID: 27535481 PMCID: PMC5002908 DOI: 10.14814/phy2.12891] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 07/18/2016] [Indexed: 11/24/2022] Open
Abstract
In women, progesterone suppresses luteinizing hormone (LH) (gonadotropin‐releasing hormone) pulse frequency, but how rapidly this occurs is unknown. In estradiol‐pretreated women in the late follicular phase, progesterone administration at 1800 did not slow sleep‐associated LH pulse frequency. However, mechanisms controlling LH pulse frequency may differ according to sleep status; and we thus hypothesized that progesterone acutely suppresses waking LH pulse frequency. This was a randomized, double‐blind, crossover study of LH secretory responses to progesterone versus placebo administered at 0600. We studied 12 normal women in the late follicular phase (cycle days 7–11), pretreated with 3 days of transdermal estradiol (0.2 mg/day). Subjects underwent a 24‐h blood sampling protocol (starting at 2000) and received either 100 mg oral micronized progesterone or placebo at 0600. In a subsequent menstrual cycle, subjects underwent an identical protocol except that oral progesterone was exchanged for placebo or vice versa. Changes in 10‐h LH pulse frequency were similar between progesterone and placebo. However, mean LH, LH pulse amplitude, and mean follicle‐stimulating hormone exhibited significantly greater increases with progesterone. Compared to our previous study (progesterone administered at 1800), progesterone administration at 0600 was associated with a similar increase in mean LH, but a less pronounced increase in LH pulse amplitude. We conclude that, in estradiol‐pretreated women in the late follicular phase, a single dose of progesterone does not suppress waking LH pulse frequency within 12 h, but it acutely amplifies mean LH and LH pulse amplitude – an effect that may be influenced by sleep status and/or time of day.
Collapse
Affiliation(s)
- Eleanor G Hutchens
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia Center for Research in Reproduction, University of Virginia School of Medicine, Charlottesville, Virginia
| | | | - Louisa C Howard
- College and Graduate School of Arts and Sciences, University of Virginia, Charlottesville, Virginia
| | - Michelle Y Abshire
- Center for Research in Reproduction, University of Virginia School of Medicine, Charlottesville, Virginia
| | - James T Patrie
- Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Christopher R McCartney
- Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia Center for Research in Reproduction, University of Virginia School of Medicine, Charlottesville, Virginia
| |
Collapse
|
38
|
Levels and actions of neuroactive steroids in the nervous system under physiological and pathological conditions: Sex-specific features. Neurosci Biobehav Rev 2016; 67:25-40. [DOI: 10.1016/j.neubiorev.2015.09.023] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 09/15/2015] [Accepted: 09/16/2015] [Indexed: 01/21/2023]
|
39
|
Giatti S, Melcangi RC, Pesaresi M. The other side of progestins: effects in the brain. J Mol Endocrinol 2016; 57:R109-26. [PMID: 27339142 DOI: 10.1530/jme-16-0061] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 06/22/2016] [Indexed: 01/06/2023]
Abstract
Progestins are a broad class of progestational agents widely differing in their chemical structures and pharmacological properties. Despite emerging data suggest that progestins, besides their action as endometrial protection, can also have multiple nonreproductive functions, much remains to be discovered regarding the actions exerted by these molecules in the nervous system. Here, we report the role exerted by different progestins, currently used for contraception or in postmenopausal hormone replacement therapies, in regulating cognitive functions as well as social behavior and mood. We provide evidence that the effects and mechanisms underlying their actions are still confusing due to the use of different estrogens and progestins as well as different doses, duration of exposure, route of administration, baseline hormonal status and age of treated women. We also discuss the emerging issue concerning the relevant increase of these substances in the environment, able to deeply affect aquatic wildlife as well as to exert a possible influence in humans, which may be exposed to these compounds via contaminated drinking water and seafood. Finally, we report literature data showing the neurobiological action of progestins and in particular their importance during neurodegenerative events. This is extremely interesting, since some of the progestins currently used in clinical practice exert neuroprotective and anti-inflammatory effects in the nervous system, opening new promising opportunities for the use of these molecules as therapeutic agents for trauma and neurodegenerative disorders.
Collapse
Affiliation(s)
- Silvia Giatti
- Department of Pharmacological and Biomolecular SciencesCenter of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Milan, Italy
| | - Roberto Cosimo Melcangi
- Department of Pharmacological and Biomolecular SciencesCenter of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Milan, Italy
| | - Marzia Pesaresi
- Department of Pharmacological and Biomolecular SciencesCenter of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
40
|
Wang C, Liu D, Chen W, Ge W, Hong W, Zhu Y, Chen SX. Progestin increases the expression of gonadotropins in pituitaries of male zebrafish. J Endocrinol 2016; 230:143-56. [PMID: 27113852 PMCID: PMC4938713 DOI: 10.1530/joe-16-0073] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 04/22/2016] [Indexed: 01/15/2023]
Abstract
Our previous study showed that the in vivo positive effects of 17α,20β-dihydroxy-4-pregnen-3-one (DHP), the major progestin in zebrafish, on early spermatogenesis was much stronger than the ex vivo ones, which may suggest an effect of DHP on the expression of gonadotropins. In our present study, we first observed that fshb and lhb mRNA levels in the pituitary of male adult zebrafish were greatly inhibited by 3 weeks exposure to 10nM estradiol (E2). However, an additional 24h 100nM DHP exposure not only reversed the E2-induced inhibition, but also significantly increased the expression of fshb and lhb mRNA. These stimulatory effects were also observed in male adult fish without E2 pretreatment, and a time course experiment showed that it took 24h for fshb and 12h for lhb to respond significantly. Because these stimulatory activities were partially antagonized by a nuclear progesterone receptor (Pgr) antagonist mifepristone, we generated a Pgr-knockout (pgr(-/-)) model using the TALEN technique. With and without DHP in vivo treatment, fshb and lhb mRNA levels of pgr(-/-) were significantly lower than those of pgr(+/+) Furthermore, ex vivo treatment of pituitary fragments of pgr(-/-) with DHP stimulated lhb, but not fshb mRNA expression. Results from double-colored fluorescent in situ hybridization showed that pgr mRNA was expressed only in fshb-expressing cells. Taken together, our results indicated that DHP participated in the regulation of neuroendocrine control of reproduction in male zebrafish, and exerted a Pgr-mediated direct stimulatory effect on fshb mRNA at pituitary level.
Collapse
Affiliation(s)
- Cuili Wang
- State Key Laboratory of Marine Environmental ScienceCollege of Ocean and Earth Sciences, Xiamen University, Xiamen, China Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological ResourcesXiamen, China
| | - Dongteng Liu
- State Key Laboratory of Marine Environmental ScienceCollege of Ocean and Earth Sciences, Xiamen University, Xiamen, China Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological ResourcesXiamen, China
| | - Weiting Chen
- Centre of ReproductionDevelopment and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Wei Ge
- Centre of ReproductionDevelopment and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Wanshu Hong
- State Key Laboratory of Marine Environmental ScienceCollege of Ocean and Earth Sciences, Xiamen University, Xiamen, China Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological ResourcesXiamen, China
| | - Yong Zhu
- State Key Laboratory of Marine Environmental ScienceCollege of Ocean and Earth Sciences, Xiamen University, Xiamen, China Department of BiologyEast Carolina University, Greenville, North Carolina, USA
| | - Shi X Chen
- State Key Laboratory of Marine Environmental ScienceCollege of Ocean and Earth Sciences, Xiamen University, Xiamen, China Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological ResourcesXiamen, China State-Province Joint Engineering Laboratory of Marine Bioproducts and TechnologyXiamen University, Xiamen, China
| |
Collapse
|
41
|
Moore AM, Campbell RE. The neuroendocrine genesis of polycystic ovary syndrome: A role for arcuate nucleus GABA neurons. J Steroid Biochem Mol Biol 2016; 160:106-17. [PMID: 26455490 DOI: 10.1016/j.jsbmb.2015.10.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 08/25/2015] [Accepted: 10/02/2015] [Indexed: 12/12/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent and distressing endocrine disorder lacking a clearly identified aetiology. Despite its name, PCOS may result from impaired neuronal circuits in the brain that regulate steroid hormone feedback to the hypothalamo-pituitary-gonadal axis. Ovarian function in all mammals is controlled by the gonadotropin-releasing hormone (GnRH) neurons, a small group of neurons that reside in the pre-optic area of the hypothalamus. GnRH neurons drive the secretion of the gonadotropins from the pituitary gland that subsequently control ovarian function, including the production of gonadal steroid hormones. These hormones, in turn, provide important feedback signals to GnRH neurons via a hormone sensitive neuronal network in the brain. In many women with PCOS this feedback pathway is impaired, resulting in the downstream consequences of the syndrome. This review will explore what is currently known from clinical and animal studies about the identity, relative contribution and significance of the individual neuronal components within the GnRH neuronal network that contribute to the pathophysiology of PCOS. We review evidence for the specific neuronal pathways hypothesised to mediate progesterone negative feedback to GnRH neurons, and discuss the potential mechanisms by which androgens may evoke disruptions in these circuits at different developmental time points. Finally, this review discusses data providing compelling support for disordered progesterone-sensitive GABAergic input to GnRH neurons, originating specifically within the arcuate nucleus in prenatal androgen induced forms of PCOS.
Collapse
Affiliation(s)
- Aleisha M Moore
- Centre for Neuroendocrinology and Department of Physiology, School of Medical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Rebecca E Campbell
- Centre for Neuroendocrinology and Department of Physiology, School of Medical Sciences, University of Otago, Dunedin 9054, New Zealand.
| |
Collapse
|
42
|
Abstract
The progesterone receptor (PGR) is a ligand-activated transcription factor with key roles in the regulation of female fertility. Much has been learned of the actions of PGR signaling through the use of pharmacologic inhibitors and genetic manipulation, using mouse mutagenesis. Characterization of rats with a null mutation at the Pgr locus has forced a reexamination of the role of progesterone in the regulation of the female reproductive cycle. We generated two Pgr mutant rat models, using genome editing. In both cases, deletions yielded a null mutation resulting from a nonsense frame-shift and the emergence of a stop codon. Similar to Pgr null mice, Pgr null rats were infertile because of deficits in sexual behavior, ovulation, and uterine endometrial differentiation. However, in contrast to the reported phenotype of female mice with disruptions in Pgr signaling, Pgr null female rats exhibit robust estrous cycles. Cyclic changes in vaginal cytology, uterine histology, serum hormone levels, and wheel running activity were evident in Pgr null female rats, similar to wild-type controls. Furthermore, exogenous progesterone treatment inhibited estrous cycles in wild-type female rats but not in Pgr-null female rats. As previously reported, pharmacologic antagonism supports a role for PGR signaling in the regulation of the ovulatory gonadotropin surge, a result at variance with experimentation using genetic ablation of PGR signaling. To conclude, our findings in the Pgr null rat challenge current assumptions and prompt a reevaluation of the hormonal control of reproductive cyclicity.
Collapse
|
43
|
Endo N, Tamesaki C, Ohkura S, Wakabayashi Y, Matsui H, Tanaka A, Watanabe T, Okamura H, Tanaka T. Differential changes in luteinizing hormone secretion after administration of the investigational metastin/kisspeptin analog TAK-683 in goats. Anim Reprod Sci 2015; 159:87-93. [DOI: 10.1016/j.anireprosci.2015.05.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 05/22/2015] [Accepted: 05/26/2015] [Indexed: 10/23/2022]
|
44
|
Overturf MD, Huggett DB. Responses to various exposure durations of levonorgestrel during early-life stages of fathead minnows (Pimephales promelas). AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2015; 161:33-40. [PMID: 25661708 DOI: 10.1016/j.aquatox.2015.01.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 01/26/2015] [Accepted: 01/28/2015] [Indexed: 06/04/2023]
Abstract
Pharmaceuticals are routinely detected in the environment; and several of these compounds have been extensively researched due to their potential impacts to the endocrine system of aquatic organisms. The negative reproductive consequences of synthetic progestins in teleost species have only recently been investigated. The current study examined different exposure periods that may be most sensitive for levonorgestrel (LNG) in early-life stages of fathead minnow larvae. Larvae were exposed to a single concentration of LNG (125ng/L) for different durations from fertilized egg through 28 days post hatch (dph) with growth and mRNA expression of FSH, 3β-HSD, 20β-HSD, and CYP19a1 measured. Regardless of the duration of exposure, LNG significantly decreased growth in the fathead minnow larvae at day 28. For both 20β-HSD and CYP19a1, mRNA expression was decreased following LNG exposure durations ≥7 days. 3β-HSD and FSH showed similar trends after exposure to LNG with later stages of development exhibiting decreased expression. 20β-HSD and 3β-HSD were the only transcripts to remain down regulated once larvae were moved to clean water after the 7-14dph LNG exposure. This study is the first to investigate the effects of exposure time to a synthetic progestin on developing fish. Future research is needed to understand what impacts these changes have on adult stages of development.
Collapse
Affiliation(s)
- Matthew D Overturf
- Department of Biology, University of North Texas, Denton, TX 76203, USA.
| | - Duane B Huggett
- Department of Biology, University of North Texas, Denton, TX 76203, USA
| |
Collapse
|
45
|
Eghlidi DH, Urbanski HF. Effects of Age and Estradiol on Gene Expression in the Rhesus Macaque Hypothalamus. Neuroendocrinology 2015; 101:236-45. [PMID: 25765287 PMCID: PMC4475460 DOI: 10.1159/000381063] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 02/18/2015] [Indexed: 11/19/2022]
Abstract
BACKGROUND The hypothalamus plays a key role in mediating the effects of estrogen on many physiological functions, including reproduction, metabolism, and thermoregulation. We have previously observed marked estrogen-dependent gene expression changes within the hypothalamus of rhesus macaques during aging, especially in the KNDy neurons of the arcuate-median eminence (ARC-ME) that produce kisspeptin, neurokinin B, and dynorphin A. Little is known, however, about the mechanisms involved in mediating the feedback from estrogen onto these neurons. METHODS We used quantitative real-time PCR to profile age- and estrogen-dependent gene expression changes in the rhesus macaque hypothalamus. Our focus was on genes that encode steroid receptors (ESR1, ESR2, PGR, and AR) and on enzymes that contribute to the local synthesis of 17β-estradiol (E2; STS, HSD3B1/2, HSD17B5, and CYP19A). In addition, we used RT(2) Profiler™ PCR Arrays to profile a larger set of genes that are integral to hypothalamic function. RESULTS KISS1, KISS1R, TAC3, and NPY2R mRNA levels increased in surgically menopausal (ovariectomized) old females relative to age-matched ovariectomized animals that received E2 hormone therapy. In contrast, PGR, HSD17B, GNRH2, SLC6A3, KISS1, TAC3, and NPY2R mRNA levels increased after E2 supplementation. CONCLUSION The rhesus macaque ARC-ME expresses many genes that are responsive to changes in circulating estrogen levels, even during old age, and these may contribute to causing the normal and pathophysiological changes that occur during menopause.
Collapse
Affiliation(s)
- Dominique H. Eghlidi
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oreg., USA
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, Oreg., USA
| | - Henryk F. Urbanski
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oreg., USA
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, Oreg., USA
- Deptartment of Physiology and Pharmacology, Oregon Health and Science University, Portland, Oreg., USA
| |
Collapse
|
46
|
Enhancement of a robust arcuate GABAergic input to gonadotropin-releasing hormone neurons in a model of polycystic ovarian syndrome. Proc Natl Acad Sci U S A 2014; 112:596-601. [PMID: 25550522 DOI: 10.1073/pnas.1415038112] [Citation(s) in RCA: 147] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Polycystic ovarian syndrome (PCOS), the leading cause of female infertility, is associated with an increase in luteinizing hormone (LH) pulse frequency, implicating abnormal steroid hormone feedback to gonadotropin-releasing hormone (GnRH) neurons. This study investigated whether modifications in the synaptically connected neuronal network of GnRH neurons could account for this pathology. The PCOS phenotype was induced in mice following prenatal androgen (PNA) exposure. Serial blood sampling confirmed that PNA elicits increased LH pulse frequency and impaired progesterone negative feedback in adult females, mimicking the neuroendocrine abnormalities of the clinical syndrome. Imaging of GnRH neurons revealed greater dendritic spine density that correlated with increased putative GABAergic but not glutamatergic inputs in PNA mice. Mapping of steroid hormone receptor expression revealed that PNA mice had 59% fewer progesterone receptor-expressing cells in the arcuate nucleus of the hypothalamus (ARN). To address whether increased GABA innervation to GnRH neurons originates in the ARN, a viral-mediated Cre-lox approach was taken to trace the projections of ARN GABA neurons in vivo. Remarkably, projections from ARN GABAergic neurons heavily contacted and even bundled with GnRH neuron dendrites, and the density of fibers apposing GnRH neurons was even greater in PNA mice (56%). Additionally, this ARN GABA population showed significantly less colocalization with progesterone receptor in PNA animals compared with controls. Together, these data describe a robust GABAergic circuit originating in the ARN that is enhanced in a model of PCOS and may underpin the neuroendocrine pathophysiology of the syndrome.
Collapse
|
47
|
Fergani C, Routly JE, Jones DN, Pickavance LC, Smith RF, Dobson H. Activation of cells containing estrogen receptor alpha or somatostatin in the medial preoptic area, arcuate nucleus, and ventromedial nucleus of intact ewes during the follicular phase, and alteration after lipopolysaccharide. Biol Reprod 2014; 91:141. [PMID: 25320149 DOI: 10.1095/biolreprod.114.122408] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Cells in the medial preoptic area (mPOA), arcuate nucleus (ARC), and ventromedial nucleus (VMN) that possess estrogen receptor alpha (ER alpha) mediate estradiol feedback to regulate endocrine and behavioral events during the estrous cycle. A percentage of ER alpha cells located in the ARC and VMN express somatostatin (SST) and are activated in response to estradiol. The aims of the present study were to investigate the location of c-Fos, a marker for activation, in cells containing ER alpha or SST at various times during the follicular phase and to determine whether lipopolysaccharide (LPS) administration, which leads to disruption of the luteinizing hormone (LH) surge, is accompanied by altered ER alpha and/or SST activation patterns. Follicular phases were synchronized with progesterone vaginal pessaries, and control animals were killed at 0, 16, 31, and 40 h (n = 4-6/group) after progesterone withdrawal (PW [time 0]). At 28 h, other animals received LPS (100 ng/kg) and were subsequently killed at 31 h or 40 h (n = 5/group). Hypothalamic sections were immunostained for c-Fos and ER alpha or SST. LH surges occurred only in control ewes with onset at 36.7 ± 1.3 h after PW; these animals had a marked increase in the percentage of ER alpha cells that colocalized c-Fos (%ER alpha/c-Fos) in the ARC and mPOA from 31 h after PW and throughout the LH surge. In the VMN, %ER alpha/c-Fos was higher in animals that expressed sexual behavior than in those that did not. SST cell activation in the ARC and VMN was greater during the LH surge than in other stages in the follicular phase. At 31 or 40 h after PW (i.e., 3 or 12 h after treatment, respectively), LPS decreased %ER alpha/c-Fos in the ARC and the mPOA, but there was no change in the VMN compared to that in controls. The %SST/c-Fos increased in the VMN at 31 h after PW (i.e., 3 h after LPS) with no change in the ARC compared to controls. These results indicate that there is a distinct temporal pattern of ER alpha cell activation in the hypothalamus during the follicular phase, which begins in the ARC and mPOA at least 6-7 h before the LH surge onset and extends to the VMN after the onset of sexual behavior and LH surge. Furthermore, during the surge, some of these ER alpha-activated cells may be SST-secreting cells. This pattern is markedly altered by LPS administered during the late follicular phase, indicating that the disruptive effects of this stressor are mediated by suppressing ER alpha cell activation at the level of the mPOA and ARC and enhancing SST cell activation in the VMN, leading to the attenuation of the LH surge.
Collapse
Affiliation(s)
- Chrysanthi Fergani
- School of Veterinary Science, University of Liverpool, Leahurst Campus, Neston, Wirral, United Kingdom
| | - Jean E Routly
- School of Veterinary Science, University of Liverpool, Leahurst Campus, Neston, Wirral, United Kingdom
| | - David N Jones
- School of Veterinary Science, University of Liverpool, Leahurst Campus, Neston, Wirral, United Kingdom
| | - Lucy C Pickavance
- School of Veterinary Science, University of Liverpool, Leahurst Campus, Neston, Wirral, United Kingdom
| | - Robert F Smith
- School of Veterinary Science, University of Liverpool, Leahurst Campus, Neston, Wirral, United Kingdom
| | - Hilary Dobson
- School of Veterinary Science, University of Liverpool, Leahurst Campus, Neston, Wirral, United Kingdom
| |
Collapse
|
48
|
Bouchard P. Selective progesterone receptor modulators: a class with multiple actions and applications in reproductive endocrinology, and gynecology. Gynecol Endocrinol 2014; 30:683-4. [PMID: 25242338 DOI: 10.3109/09513590.2014.950647] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Among the remarkable progress made in the treatment of hormone-dependent diseases, recently selective steroid receptor modulators (SPRM) have been remarkable tools to improve the prognosis of multiple hormone-dependent cancers (breast, prostate). Because of their remarkable properties, both agonistic and mainly antagonistic, to modulate progesterone action. They have allowed a better understanding of progesterone action and progress in contraception and treatment of abnormal uterine bleeding as well as in the medical treatment of uterine fibroids.
Collapse
|
49
|
Adib A, Freret S, Touze JL, Lomet D, Lardic L, Chesneau D, Estienne A, Papillier P, Monniaux D, Pellicer-Rubio MT. Progesterone improves the maturation of male-induced preovulatory follicles in anoestrous ewes. Reproduction 2014; 148:403-16. [DOI: 10.1530/rep-14-0263] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The first ovulation induced by male effect in sheep during seasonal anoestrus usually results in the development of a short cycle that can be avoided by progesterone priming before ram introduction. In elucidating the involvement of the hypothalamic–pituitary–gonadal axis in the occurrence of short cycles, the effects of progesterone and the time of anoestrus on the development of male-induced preovulatory follicles were investigated in anoestrous ewes using morphological, endocrine and molecular approaches. Ewes were primed with progesterone for 2 (CIDR2) or 12 days (CIDR12) and untreated ewes used as controls during early (April) and late (June) anoestrus. The duration of follicular growth and the lifespan of the male-induced preovulatory follicles were prolonged by ∼1.6 days in CIDR12 ewes compared with the controls. These changes were accompanied by a delay in the preovulatory LH and FSH surges and ovulation. Intra-follicular oestradiol concentration and mRNA levels ofLHCGRandSTARin the granulosa and theca cells of the preovulatory follicles were higher in CIDR12 ewes than the control ewes. The expression of mRNA levels ofCYP11A1andCYP17A1also increased in theca cells of CIDR12 ewes. CIDR2 ewes gave intermediate results. Moreover, ewes ovulated earlier in June than in April, without changes in the duration of follicular growth, but these effects were unrelated to the lifespan of corpus luteum. Our results give the first evidence supporting the positive effect of progesterone priming on the completion of growth and maturation of preovulatory follicles induced by male effect in seasonal anoestrous ewes, thereby preventing short cycles.
Collapse
|
50
|
Naugle MM, Nguyen LT, Merceron TK, Filardo E, Janssen WGM, Morrison JH, Rapp PR, Gore AC. G-protein coupled estrogen receptor, estrogen receptor α, and progesterone receptor immunohistochemistry in the hypothalamus of aging female rhesus macaques given long-term estradiol treatment. ACTA ACUST UNITED AC 2014; 321:399-414. [PMID: 24862737 DOI: 10.1002/jez.1871] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 04/28/2014] [Indexed: 12/15/2022]
Abstract
Steroid hormone receptors are widely and heterogeneously expressed in the brain, and are regulated by age and gonadal hormones. Our goal was to quantify effects of aging, long-term estradiol (E2 ) treatment, and their interactions, on expression of G protein-coupled estrogen receptor (GPER), estrogen receptor α (ERα) and progesterone receptor (PR) immunoreactivity in two hypothalamic regions, the arcuate (ARC) and the periventricular area (PERI) of rhesus monkeys as a model of menopause and hormone replacement. Ovariectomized (OVX) rhesus macaques were young (∼ 11 years) or aged (∼ 25 years), given oil (vehicle) or E2 every 3 weeks for 2 years. Immunohistochemistry and stereologic analysis of ERα, PR, and GPER was performed. More effects were detected for GPER than the other two receptors. Specifically, GPER cell density in the ARC and PERI, and the percent of GPER-immunoreactive cells in the PERI, were greater in aged than in young monkeys. In addition, we mapped the qualitative distribution of GPER in the monkey hypothalamus and nearby regions. For ERα, E2 treated monkeys tended to have higher cell density than vehicle monkeys in the ARC. The percent of PR density in the PERI tended to be higher in E2 than vehicle monkeys of both ages. This study shows that the aged hypothalamus maintains expression of hormone receptors with age, and that long-term cyclic E2 treatment has few effects on their expression, although GPER was affected more than ERα or PR. This result is surprising in light of evidence for E2 regulation of the receptors studied here, and differences may be due to the selected regions, long-term nature of E2 treatment, among other possibilities.
Collapse
Affiliation(s)
- Michelle M Naugle
- Institute for Neuroscience, University of Texas at Austin, Austin, Texas
| | | | | | | | | | | | | | | |
Collapse
|